PROSTATE CANCER-ASSOCIATED SECRETED PROTEINS

-

Methods are provided for treating a subject with prostate cancer and/or diagnosing a subject at risk for prostate cancer, which can include measuring increased expression of at least two prostate cancer-related molecules in a sample obtained from a subject, including the prostate cancer-related molecules AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4. The methods can include administering at therapy to a subject with prostate cancer. Methods are provided for treating a subject with intermediate- or high-risk prostate cancer, which can include measuring increased expression of MMP9 in a sample obtained from a subject compared to a control representing expression of MMP9 expected in a sample from a subject who has low-risk prostate cancer.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATION

This application claims the benefit of the earlier filing date of U.S. Provisional Application No. 62/432,946, filed Dec. 12, 2016, which is incorporated by reference in its entirety.

ACKNOWLEDGMENT OF GOVERNMENT SUPPORT

This invention was made with government support under U01-CA111244, awarded by the National Institute of Health; under W81XWH-16-1-0614 awarded by the Department of Defense Prostate Cancer Research Program; under U01CA86402 awarded by the National Cancer Institute Early Detection Research Network Clinical Epidemiology Center; under P41GM103493 awarded by the National Institute of General Medical Sciences Biomedical Technology Research Resource; and under DE-AC05-76RL01830 awarded by the Department of Energy. The government has certain rights in the invention.

FIELD

This application provides methods of diagnosing and/or treating prostate cancer, following determining expression levels of several prostate cancer-related secretory molecules, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4, for example in a urine sample.

PARTIES TO JOINT RESEARCH AGREEMENT

This application describes and claims certain subject matter that was developed under a written joint research agreement between Battelle Memorial Institute and the University of Washington.

BACKGROUND

Prostate cancer is the one of the most common cancers diagnosed in the United States. A prostate cancer diagnosis can vary from a low- or intermediate-risk to a high-risk diagnosis, which means that the patient has a low-, intermediate-, or high-risk of pathological and biochemical outcomes after treatment (e.g., by radical prostatectomy); metastasis; prostate cancer-specific mortality; and all-cause mortality. A diagnosis of a particular risk category can determine the course of treatment, which can, for example, range from monitoring, in the case of low-risk cancers, to radiation or surgical procedures for higher risk cancers.

Common methods of screening for prostate cancer include a digital rectal exam (DRE) and serum prostate-specific antigen (sPSA) screening, which can be administered alone or in combination. The use of DRE is not favored by patients, and sPSA testing is controversial. One prostate cancer urine test measures the cancer-specific non-coding transcript PCA3 from released prostate cancer cells, but only has a sensitivity of 65% and a specificity of 66%.

Thus, while screening men for prostate cancer decreases mortality from the disease, a need remains for a more accurate and non-invasive means of screening for prostate cancer. There is also a need for an accurate, non-invasive means of distinguishing prostate cancer in different risk categories with the concern of overtreatment. For example, low-risk prostate cancer patients can remain healthy for a long time without undergoing invasive and painful surgery with potentially harmful side effects, and, thus, are considered ideal candidates for observation-based therapies, such as watchful waiting and active surveillance.

SUMMARY

It is shown herein that a combination of prostate cancer-related molecules, such as two or more of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4, can be detected in patient urine samples (such as urine from a human subject) and can be used as biomarkers to diagnose and/or treat a patient with or at risk for prostate cancer. Further, it is demonstrated that biomarker MMP9 can distinguish with significant accuracy subjects with low-risk prostate cancer from subjects with intermediate- or high-risk prostate cancer.

Methods are provided for treating a subject with prostate cancer. Such methods can include measuring expression of at least two prostate cancer-related molecules in a sample obtained from a subject, including at least two of (such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 of) the prostate cancer-related molecules AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4. In some examples, expression levels are normalized to PSA levels detected in urine. The methods can further include measuring increased expression of the at least two prostate cancer-related molecules, for example as compared to a control or reference value representing expression for each of the at least two prostate cancer-related molecules expected in a sample from a subject who does not have prostate cancer (e.g., as compared to a threshold of expression of any of these molecules established from a subject or subjects, such as a cohort of control subjects). In addition, the methods can include administering at least one of watchful waiting, active surveillance, surgery, radiation, hormone therapy, chemotherapy, brachytherapy, cryotherapy, ultrasound, bisphosphate therapy, biologic therapy, or vaccine therapy to the subject with prostate cancer, thereby treating the subject.

Methods are provided for diagnosing prostate cancer in a subject. The methods can include detecting the expression of at least two prostate cancer-related molecules in a sample obtained from a subject, including at least two of (such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 of) the prostate cancer-related molecules AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4. In some examples, expression levels are normalized to PSA levels detected in urine. The methods can further include comparing the expression of the at least two prostate cancer-related molecules in the sample obtained from the subject to at least one control or reference value representing expression for each of the at least two prostate cancer-related molecules expected in a sample from a subject who does not have prostate cancer (e.g., as compared to a threshold of expression of any of these molecules established from a subject or subjects, such as a cohort of control subjects). In addition, the methods can include determining that the subject has prostate cancer when increased expression of the at least two prostate cancer-related molecules between the sample and the control is detected.

In some examples, the at least two prostate cancer-related molecules can include all of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4; the combinations of prostate cancer-related molecules listed in FIG. 15; low-abundance molecules, such as AGR2, AGR3, CCL3, CEACAM5, and CEACAM6; or moderate-to-low-abundance molecules, such as CRISP3, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4.

Methods are provided for treating a subject with intermediate- or high-risk prostate cancer. The methods can include measuring expression of MMP9 in a sample obtained from a subject, which can be determined based on MMP9 protein concentration using at least one surrogate peptide of MMP9, and the surrogate peptide can be at least one of FQTFEGDLK (SEQ ID NO: 41) or LGLGADVAQVTGALR (SEQ ID NO: 42). In some examples, the MMP9 expression level is normalized to PSA levels detected in urine. The methods further include measuring increased expression of MMP9 in the sample obtained from the subject as compared to a control or reference value representing expression of MMP9 expected in a sample from a subject who has low-risk prostate cancer (e.g., as compared to a threshold of expression of MMP9 established from a subject or subjects, such as a cohort of control subjects). In addition, the methods can include administering treatment for intermediate- or high-risk prostate cancer, thereby treating the subject.

Methods are provided for diagnosing intermediate- or high-risk prostate cancer. The methods can include detecting expression of MMP9 in a sample obtained from a subject, which can be determined based on MMP9 protein concentration using at least one surrogate peptide of MMP9, and the surrogate peptide can be at least one of FQTFEGDLK (SEQ ID NO: 41) or LGLGADVAQVTGALR (SEQ ID NO: 42). In some examples, expression levels are normalized to PSA levels detected in urine. In further examples, the methods can include comparing MMP9 expression in the sample obtained from the subject to MMP9 expression expected in a sample from a subject who has low-risk prostate cancer (e.g., a reference value representing MMP9 expression expected in a subject with low-risk prostate cancer, such as a threshold of expression of MMP9 established from a subject or subjects, such as a cohort of control subjects). In addition, the methods can include determining that the subject has intermediate- or high-risk prostate cancer when increased expression of MMP9 between the sample and the control is detected.

In some examples, the methods can include determining expression based on protein concentration, which can be determined using the concentration of at least one surrogate peptide of the protein, such as a peptide listed in FIG. 4. In other examples, the methods can include determining the protein concentration using an immunoassay, such as an ELISA. In additional examples, the protein concentration can be determined using mass spectrometry, for example, using LC-SRM, LG-SRM, or PRISM-SRM.

In some other examples, the methods include normalizing expression of the at least two prostate cancer-related molecules to the amount of a prostate protein, such as PSA, which can be determined using, for example, at least one surrogate peptide of PSA, such as IVGGWECEK (SEQ ID NO: 70) or LSEPAELTDAVK (SEQ ID NO: 71).

In additional examples, the sample can be a urine sample, and, in further examples, the subject can be a human subject. In particular examples, the expression detected can have an AUC value of greater than 0.8, such as at least 0.85, at least 0.9, or at least 0.95. In particular examples, the disclosed methods have a sensitivity of at least 78 to 86% and specificity of at least 100%.

Also provided are methods that include treating a sample (e.g., urine sample) obtained from a subject (such as one known or suspected to have prostate cancer) with a protease and measuring expression of AGR2, AGR3, CEACAM5, CD90, and SFRP4, for example using mass spectrometry. In some examples, the methods also includes measuring expression of CRISPS, CCL3, CEACAM6, IL24, MMP9, CXCL14, and POSTN, for example using mass spectrometry. In some examples, one or more of the surrogate peptides disclosed herein (e.g., those in FIGS. 2, 4 and 15) are used to measure expression of these proteins, for example using mass spectrometry.

The foregoing and other objects and feature of the disclosure will become more apparent from the following detailed description, which proceeds with reference to the accompanying figures.

BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

FIG. 1 shows a panel that lists the query result from the database UrinePA. The parameter “observed” indicates abundance.

FIG. 2 shows thirteen prostate cancer-associated secreted proteins and their surrogate peptides. For each surrogate peptide, the three best transitions without co-eluting interference were monitored. The “a” indicates that endogenous light peptides were detected in the pooled urine sample. The “b” indicates that cysteine was synthesized as carbamidomethyl cysteine. Peptides shown from top to bottom are SEQ ID NOS: 6-9, 18-20, 72-74, 23-26, 29-31, 12-15, 47-50, 34-37, 40-44, 58-61, 64-67, 53-55, and 75-78.

FIGS. 3A-3B show extracted ion chromatograms (XICs) of detected proteins in a single urine sample, P07-031C. Seven proteins (CD90, CRISP3, CXCL14, IL24, MMP9, POSTN, and SFRP4) were detected by LG-SRM, and the other five (AGR2, AGR3, CCL3, CEACAM5, and CEACAM6) were in extremely low abundance and were detected by PRISM-SRM. The monitored transitions for the surrogate peptides of each protein are THY1/CD90: VLYLSAFTSK (SEQ ID NO: 53), 564.8/916.5 (blue), 564.8/640.3 (chestnut), and 564.8/753.4 (purple); CRISP3: WANQCcamNYR (SEQ ID NO: 12), 556.2/854.3 (purple), 556.2/925.4 (blue), and 556.2/612.3 (chestnut); CXCL14: MVIITTK (SEQ ID NO: 47), 403.2/575.4 (purple), 403.2/674.4 (blue), and 403.2/462.3 (chestnut); IL24: LWEAFWAVK (SEQ ID NO: 34), 575.3/850.4 (blue), 575.3/721.4 (purple), and 575.3/650.4 (chestnut); MMP9: AVIDDAFAR (SEQ ID NO: 40), 489.3/807.4 (blue), 489.3/694.3 (purple), and 489.3/579.3 (chestnut); POSTN:

AAAITSDILEALGR (SEQ ID NO: 58), 700.9/1074.8 (blue), 700.9/973.5 (purple), and 700.9/771.5 (chestnut); SFRP4: GVCcamISPEAIVTDLPEDVK (SEQ ID NO: 64), 971.5/587.3 (chestnut), 971.5/916.5 (blue), and 971.5/1425.7 (purple); AGR2: LPQTLSR (SEQ ID NO: 3), 407.7/351.2 (chestnut), 407.7/476.2 (purple), and 407.7/604.3 (blue); AGR3: LYTYEPR (SEQ ID NO: 6), 471.2/665.3 (blue), 471.2/272.2 (purple), and 471.2/277.2 (chestnut); CCL3: QVCcamADPSEEWVQK (SEQ ID NO: 16), 788.4/1002.5 (chestnut), 788.4/1117.5 (purple), and 788.4/1188.6 (blue); CEACAM5: SDLVNEEATGQFR (SEQ ID NO: 23), 733.3/679.4 (blue), 733.3/1051.5 (chestnut), and 733.3/937.4 (purple); CEACAM6: SDPVTLNVLYGPDGPTISPSK (SEQ ID NO: 30), 1079.1/1055.5 (blue), 1079.1/331.2 (chestnut), and 1079.1/998.5 (purple).

FIG. 4 shows prostate cancer-associated secreted proteins and their surrogate peptides. The “a” indicates that these surrogate peptides were confidently detected in the pooled urine sample. The “b” indicates that the cysteine was synthesized as carbamidomethyl cysteine. Peptides shown from top to bottom are SEQ ID NOS: 3, 6, 18, 23-25, 29-30, 12-13, 47-48, 34, 40-43, 58, 64, and 53-55.

FIGS. 5A-5F show correlation plots between any two MMP9 surrogate peptides in 20 urine samples. FIG. 5A shows a relative abundance correlation between FQTFEGDLK (y-axis; SEQ ID NO: 41) and AVIDDAFAR (x-axis; SEQ ID NO: 40); FIG. 5B shows a relative abundance correlation between LGLGADVAQVTGALR (y-axis; SEQ ID NO: 42) and AVIDDAFAR (x-axis; SEQ ID NO: 40); FIG. 5C shows a relative abundance correlation between SLGPALLLLQK (SEQ ID NO: 43) and AVIDDAFAR (SEQ ID NO: 40); FIG. 5D shows a relative abundance correlation between SLGPALLLLQK (y-axis; SEQ ID NO: 43) and LGLGADVAQVTGALR (x-axis; SEQ ID NO: 42); FIG. 5E shows a relative abundance correlation between LGLGADVAQVTGALR (y-axis; SEQ ID NO: 42) and FQTFEGDLK (x-axis; SEQ ID NO: 41); FIG. 5F shows a relative abundance correlation between SLGPALLLLQK (y-axis; SEQ ID NO: 43) and FQTFEGDLK (y-axis; SEQ ID NO: 41). L/H=the ratio of SRM signal from endogenous peptide over heavy-labeled internal standard. R2 values range from 0.59 to 0.93.

FIGS. 6A-6B show correlation curves between two surrogate peptides from the same protein. FIG. 6A shows CD90 (with the removal of the point with the red circle, the correlation coefficient of R2 significantly decreased from 0.70 to 0.21); FIG. 6B shows CRISP3 (with the removal of the point with the red circle, the correlation coefficient of R2 significantly decreased from 0.65 to 0.14).

FIGS. 7A-7B show correlation curves between two PSA surrogate peptides in 20 urine subjects. FIG. 7A shows the entire range of relative abundance of the peptides; FIG. 7B shows a small range of relative abundance. Data points with the blue dash circle significantly deviated from the correlation curve.

FIG. 8 shows a summary of SRM measurements of PSA protein in 27 clinical urine samples including 7 post-op subjects (two purified PSA internal standards, IVGGWECcamEK (SEQ ID NO: 70) and LSEPAELTDAVK (SEQ ID NO: 71), were spiked at 1 fmol/μL and 10 fmol/μL respectively). The “a” indicates that the L/H ratios were corrected according to the correlation curve of the two PSA surrogate peptides as well as the experimental observation (P06017Pre: the measured value of 0.828 was changed into 1.656; P07040Pre: the measured value of 0.261 was changed into 0.366; P08015Pre: the measured value of 3.240 was changed into 4.739).

FIG. 9 shows extracted ion chromatograms of transitions monitored for the PSA peptide LSEPAE(L/I)TDAVK (SEQ ID NO: 71) in urine from two patients (for the two deviated data points on the correlation curve at the low concentration range in FIG. 7B).

FIG. 10 shows an estimation of the percentage of PSA from the post-op urine over PSA from the non-cancer urine (the surrogate peptide IVGGWECcamEK was used; SEQ ID NO: 70). The “a” indicates that the L/H ratios were corrected according to the correlation curve of the two PSA surrogate peptides (P07040Pre: the measured value of 0.261 was changed into 0.366; P08015Pre: the measured value of 3.240 was changed into 4.739).

FIGS. 11A-11B show a summary of multiplex SRM measurements of prostate cancer-associated secreted proteins in 20 clinical urine samples.

FIG. 12 shows the performance of surrogate peptide markers derived from 10 prostate cancer-associated secreted proteins in 20 urine samples (14 cancer and 6 non-cancer samples). The “a” indicates that P values were obtained from the Mann-Whitney U test. The “b” indicates that these are the sensitivity and specificity at the optimal cutoff point (i.e., the best sum of the sensitivity and specificity). The “c” indicates that the cysteine was synthesized as carbamidomethyl cysteine. Peptides shown from top to bottom are SEQ ID NOS: 3, 6, 12-13, 53-55, 47, 34, 40-43, and 64.

FIGS. 13A-13B show the SRM signal ratio of urinary secreted protein/PSA, i.e., (L/H)peptide marker/(L/H)PSA from SRM measurements in 20 clinical urine samples (crude internal standards for prostate cancer-associated secreted proteins and purified internal standard for PSA surrogate peptide IVGGWECcamEK were spiked at 10 fmol/μL and 1 fmol/μL, respectively; SEQ ID NO: 70). The “a” indicates that the L/H ratios were corrected according to the correlation curve of the two PSA surrogate peptides (P07040Pre: the measured value of 0.261 was changed into 0.366; P08015Pre: the measured value of 3.240 was changed into 4.739). Peptides shown from left to right in FIG. 13A are SEQ ID NOS: 3, 6, 24, 29, 12-13, and 53-54. Peptides shown from left to right in FIG. 13B are SEQ ID NOS: 47, 34, 40-43, and 64.

FIGS. 14A-14F show urine protein biomarkers for prostate cancer. FIG. 14A shows the CEACAM5 relative abundance between non-cancer (n=6) and cancer urine (n=14; P=0.322); FIG. 14B shows an ROC curve analysis of the relative abundance of CEACAM5 in the measured 20 urine samples; FIG. 14C shows CEACAM5/PSA concentration ratios between non-cancer and cancer (P=0.012); FIG. 14D shows an ROC curve analysis of the CEACAM5/PSA concentration ratios; FIG. 14E shows significant differentiation between non-cancer and cancer (P=0.0034) with the use of the best peptide combination; FIG. 14F shows an ROC curve analysis of the best peptide combination. The relative abundances of CEACAM5 and PSA were derived from their surrogate peptides, SDLVNEEATGQFR (SEQ ID NO: 23) and IVGGWECcamEK (SEQ ID NO: 70), respectively. The best peptide combination: LPQTLSR/AGR2 (SEQ ID NO: 3), LYTYEPR/AGR3 (SEQ ID NO: 6), SDLVNEEATGQFR/CEACAM5 (SEQ ID NO: 23), VTSLTACLVDQSLR/CD90 (SEQ ID NO: 54), and GVCISPEAIVTDLPEDVK/SFRP4 (SEQ ID NO: 64).

FIG. 15 shows selected combinations of multiple markers for achieving better discrimination than individual markers between cancer and non-cancer. The “a” indicates that the P values were obtained from the Mann-Whitney U test. The “b” indicates that these are the sensitivity and specificity at the optimal cutoff point (i.e., the best sum of the sensitivity and specificity). The “c” indicates that the cysteine was synthesized as carbamidomethyl cysteine. Peptides shown from top to bottom are SEQ ID NOS: 3, 6, 12-13, 23, 29, 34, 40-42, 54-55, and 64.

FIGS. 16A-16B show urinary AGR2 measured by ELISA. FIG. 16A shows an ROC plot for urinary AGR2 determined by ELISA. The P value obtained for this cohort was 0.01. FIG. 16B shows the urinary levels of AGR2 in a non-cancer healthy male (P16-050A-J), which were measured by ELISA using samples donated within a period of 14 days. Buffer was the negative control, and PC3 was the positive control. In bar 4, alcohol was added to P16-050A urine before the ELISA was performed.

FIG. 17 shows PSA concentrations in urine and serum for 20 measured subjects (urinary PSA and serum PSA concentrations were obtained from SRM measurements and ELISA measurements, respectively). The “a” indicates that the L/H ratios were corrected according to the correlation curve of the two PSA surrogate peptides (P07040Pre: the measured value of 0.261 was changed into 0.366; P08015Pre: the measured value of 3.240 was changed into 4.739).

FIGS. 18A-18B show the ratios of secreted protein over PSA concentrations, urinary PSA (uPSA) and serum PSA (sPSA) between low volume/low grade cancer (n=6) and significant cancer (n=5). The low volume/low grade cancer: Gleason score≤6 and tumor volume≤0.5 cc; the significant cancer: Gleason score>6 and tumor volume>0.5 cc. Peptides shown from left to right in FIG. 18A are SEQ ID NOS: 3, 6, 24, 29, and 12-13. Peptides shown from left to right in FIG. 18B are SEQ ID NOS: 53-55, 47, 34, 40-43, and 64.

FIGS. 19A-19C show stratification of prostate cancer based on tumor volume and Gleason score. FIG. 19A shows the relative abundance ratios of FQTFEGDLK/MMP9 over IVGGWECcamEK/PSA (SEQ ID NO: 41 and SEQ ID NO: 70) between low volume/low grade cancer (n=6) and significant cancer (n=5), P=0.022; FIG. 19B shows uPSA between low volume/low grade cancer and significant cancer (P=0.93); FIG. 19C shows a comparison of sPSA between low volume/low grade cancer and significant cancer (P=0.32).

FIGS. 20A-20B show gene expression levels of protein markers in Gleason 3 (labeled 05-179_CD26t) vs. Gleason 4 (labeled 08-032_CP_epi_CD26posi) cancer cells. Differential expression is displayed using gray scale (FIG. 20A) and histogram (FIG. 20B) formats.

SEQUENCE LISTING

The nucleic and amino acid sequences listed in the accompanying sequence listing are shown using standard letter abbreviations for nucleotide bases, and three letter code for amino acids, as defined in 37 C.F.R. 1.822. Only one strand of each nucleic acid sequence is shown, but the complementary strand is understood as included by any reference to the displayed strand. The Sequence Listing is submitted as an ASCII text file, created on December 6, 2017, 80 KB, which is incorporated by reference herein. In the accompanying sequence listing:

SEQ ID NOS: 1 and 2 are exemplary AGR2 amino acid and nucleic acid sequences, respectively.

SEQ ID NO: 3 is an amino acid sequence of an AGR2 peptide.

SEQ ID NO: 4 and 5 are exemplary AGR2 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 6-9 are exemplary ARG3 peptide sequences.

SEQ ID NOS: 10 and 11 are exemplary CRISP3 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 12-15 are exemplary CRISP3 peptide sequences.

SEQ ID NOS: 16 and 17 are exemplary CCL3 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 18-20 are exemplary CCL3 peptide sequences.

SEQ ID NOS: 21-22 are exemplary CEACAM5 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 23-26 are exemplary CEACAM5 peptide sequences.

SEQ ID NO: 27 are exemplary CEACAM6 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 29-31 are exemplary CEACAM6 peptide sequences.

SEQ ID NOS: 32-33 are exemplary IL24 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 34-37 are exemplary IL24 peptide sequences.

SEQ ID NOS: 38-39 are exemplary MMP9 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 40-44 are exemplary MMP9 peptide sequences.

SEQ ID NOS: 45-46 are exemplary CXCL14 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 47-50 are exemplary CXCL14 peptide sequences.

SEQ ID NOS: 51-52 are exemplary CD90 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 53-55 are exemplary CD90 peptide sequences.

SEQ ID NOS: 56-57 are exemplary POSTN amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 58-61 are exemplary POSTN peptide sequences.

SEQ ID NOS: 62-63 are exemplary SFRP4 amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 64-67 are exemplary SFRP4 peptide sequences.

SEQ ID NOS: 68-69 are exemplary PSA amino acid and nucleic acid sequences, respectively.

SEQ ID NOS: 70-71 are exemplary PSA peptide sequences.

SEQ ID NOS: 72-74 are exemplary CCL4 peptide sequences.

SEQ ID NOS: 75-78 are exemplary WISP peptide sequences.

DETAILED DESCRIPTION

The following explanations of terms and methods are provided to better describe the present disclosure and to guide those of ordinary skill in the art in the practice of the present disclosure. The singular forms “a,” “an,” and “the” refer to one or more than one, unless the context clearly dictates otherwise. For example, the term “comprising a protein” includes single or plural cells and is considered equivalent to the phrase “comprising at least one protein.” The term “or” refers to a single element of stated alternative elements or a combination of two or more elements, unless the context clearly indicates otherwise. As used herein, “comprises” means “includes.” Thus, “comprising A or B,” means “including A, B, or A and B,” without excluding additional elements. Dates of GenBank® Accession Nos. referred to herein are the sequences available at least as early as Dec. 12, 2016. All references and GenBank® Accession numbers cited herein are incorporated by reference in their entirety.

Unless explained otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this disclosure belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, suitable methods and materials are described below. The materials, methods, and examples are illustrative only and not intended to be limiting.

In order to facilitate review of the various embodiments of the disclosure, the following explanations of specific terms are provided.

Administration: To provide or give a subject a therapeutic intervention, such as a therapeutic drug, procedure, or protocol (e.g., for a subject with prostate cancer, docetaxel, prostatectomy, and active surveillance, respectively). Exemplary routes of administration for drug therapy include, but are not limited to, oral, injection (such as subcutaneous, intramuscular, intradermal, intraperitoneal, intratumoral, intraprostatic, and intravenous), sublingual, rectal, transdermal, intranasal, and inhalation routes.

Anterior gradient 2 (AGR2): Also known as AG2 (e.g., OMIM 606358); protein disulfide isomerase family A, member 17 (PD1A17 or member 17); or secreted cement gland protein XAG-2 homolog, AGR2 belongs to the protein disulfide isomerase (PDI) family. AGR2 plays a role in regulating the response to DNA damage as well as cell migration, growth, proliferation, and transformation. AGR2 overexpression plays a role in cancer and metastasis.

Includes AGR2 nucleic acid molecules and proteins. AGR2 sequences are publicly available. For example, GenBank® Accession Nos. NM_006408.3, NM_001106725.1, and NM_011783.2 disclose exemplary human, rat, and mouse AGR2 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_006399.1, NP_001100195.1, and NP_035913.1 disclose exemplary human, rat, and mouse AGR2 protein sequences, respectively. One of ordinary skill in the art can identify additional AGR2 nucleic acid and protein sequences, including AGR2 variants that retain AGR2 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Anterior gradient 3 (AGR3): Also known as AG3 (e.g., OMIM 609482); protein disulfide isomerase family A, member 18 (PD1A18 or member 18); or breast cancer membrane protein 11 (BCMP11), AGR3 belongs to the protein disulfide isomerase (PDI) family. AGR3 is expressed in certain epithelial and cancerous cells, such as breast and prostate cancer cells as well as cancerous epithelial cells, but exhibits restricted expression in most normal cells.

Includes AGR3 nucleic acid molecules and proteins. AGR3 sequences are publicly available. For example, GenBank® Accession Nos. NM_176813.4, NM_001106724.1, and NM_207531.3, disclose exemplary human, rat, and mouse AGR3 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_789783.1, NP_001100194.1, and NP_997414.2 disclose exemplary human, rat, and mouse AGR3 protein sequences, respectively. One of ordinary skill in the art can identify additional AGR3 nucleic acid and protein sequences, including AGR3 variants that retain AGR3 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Cluster Designation 90 (CD90): Also known as THY-1 T-cell antigen (THY1; e.g., OMIM 188230), CD90 is a cell surface glycoprotein in the immunoglobulin superfamily. CD90 is expressed by multiple cell types, including endothelial, smooth muscle, bone marrow, umbilical cord blood, fibroblasts and hemopoietic cells, and in tissues such as nervous and lymphoid tissues. Further, CD90 functions as a tumor suppressor and plays a role in cell adhesion and communication as well as immunity.

Includes CD90 nucleic acid molecules and proteins. CD90 sequences are publicly available. For example, GenBank® Accession Nos. BC065559.1, NM_012673.2, and NM_009382.3 disclose exemplary human, rat, and mouse CD90 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_001298091.1, NP_036805.1, and NP_033408.1 disclose exemplary human, rat, and mouse CD90 protein sequences, respectively. One of ordinary skill in the art can identify additional CD90 nucleic acid and protein sequences, including CD90 variants that retain CD90 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5): Also known as cluster of differentiation 66e (CD66e; e.g., OMIM 114890), CEACAM5 is an immunoreactive glycoprotein in the CEA (carcinoembryonic antigen) family. Many CEACAM family proteins are expressed in hematopoietic cells, and CEACAM5 plays a role in cell signaling, adhesion, differentiation, apoptosis, and polarity. Further, elevated levels of CEA proteins have been found in colorectal and other cancers as well as in patients with benign liver disease.

Includes CEACAM5 nucleic acid molecules and proteins. CEACAM5 sequences are publicly available. For example, GenBank® Accession Nos. NM_004363.5 and NM_028480.2 disclose exemplary human and mouse CEACAM5 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_001278413.1 and NP_082756.1 disclose exemplary human and mouse CEACAM5 protein sequences, respectively. One of ordinary skill in the art can identify additional CEACAM5 nucleic acid and protein sequences, including CEACAM5 variants that retain CEACAM5 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6): Also known as non-specific cross-reacting antigen (NCA; e.g., OMIM 163980), normal cross-reacting antigen CEA-like protein (CEAL), and cluster designation 66c (CD66c), CEACAM6 is a cell surface glycoprotein in the CEA family. CEACAM6 is expressed in neutrophils, affects tumor cell sensitivity to adenovirus infection, and is a receptor for E. coli adhesion to epithelial cells in patients with Crohn's disease. Further, CEACAM6 plays a role in platelet activation, signaling, and aggregation as well as cell surface interactions at the walls of blood vessels.

Includes CEACAM6 nucleic acid molecules and proteins. CEACAM6 sequences are publicly available. For example, GenBank® Accession Nos. NM_002483.6 and BC078962.1 disclose exemplary human and rat CEACAM6 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_002474.4 and AAH78962.1 disclose exemplary human and rat CEACAM6 protein sequences, respectively. One of ordinary skill in the art can identify additional CEACAM6 nucleic acid and protein sequences, including CEACAM6 variants that retain CEACAM6 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Chemokine (C-C Motif) ligand 3 (CCL3): Also known as small inducible cytokine A3 (SCYA3; e.g., OMIM 182283), macrophage inflammatory protein 1-α (MIP1α), and tonsillar lymphocyte LD78 α protein (LD78-α), CCL3 is a monokine involved in the acute inflammatory state of polymorphonuclear leukocyte recruitment and activation. CCL3 is expressed in many cell types, but most notably macrophages, dendritic cells, and lymphocytes. Further, CCL3 plays a key role in inflammation and the immune response to infection and can promote homeostasis.

Includes CCL3 nucleic acid molecules and proteins. CCL3 sequences are publicly available. For example, GenBank® Accession Nos. NM_002983.2, NM_013025.2, and NM_011337.2, disclose exemplary human, rat, and mouse CCL3 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_002974.1, EDM05492.1, and NP_035467.1 disclose exemplary human, rat, and mouse CCL3 protein sequences, respectively. One of ordinary skill in the art can identify additional CCL3 nucleic acid and protein sequences, including CCL3 variants that retain CCL3 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Control: A reference standard. In some embodiments, the control is a sample obtained from one or more subjects without prostate cancer (e.g., a urine sample from one or more subjects without prostate cancer). In other embodiments, the control is a sample obtained from one or more subjects without intermediate- or high-risk prostate cancer (e.g., a urine sample from one or more subjects with low-risk prostate cancer). In some embodiments, the control includes more than one subject, such as a cohort of control subjects. In still further embodiments, the control is a reference value, range of values, or threshold of values, such as from one or more subjects (e.g., a cohort). The historical control or standard (e.g., a previously tested control sample with a known prognosis or outcome or group of samples that represent baseline or normal values).

Cysteine-rich secretory protein 3 (CRISP3): Also known as CRSS and Aeg2, CRISP3 is in the cysteine-rich secretory protein subgroup of the CAP protein superfamily, the subgroup members of which are implicated in mammalian reproductive system function. CRISP3 is expressed in neutrophils, reproductive organs and glands, and the thymus and colon. CRISP3 plays a role in endometrial remodeling and repair (e.g., during the menstrual cycle and pregnancy), in prostate cancer, and immunity (e.g., in hepatitis C and Sjögren's syndrome).

Includes CRISP3 nucleic acid molecules and proteins. CRISP3 sequences are publicly available. For example, GenBank® Accession Nos. BC069602.1 and NM_009639.2 disclose exemplary human and mouse CRISP3 nucleotide sequences, respectively, and GenBank® Accession Nos. EAX04348.1 and AAI32539.1 disclose exemplary human and mouse CRISP3 protein sequences, respectively. One of ordinary skill in the art can identify additional CRISP3 nucleic acid and protein sequences, including CRISP3 variants that retain CRISP3 biological activity (such as having increased levels in urine from a subject with prostate cancer).

CXC motif, ligand 14 (CXCL14): Also known as small inducible cytokine subfamily B, member 14 (SCYB14; e.g., OMIM 604186), CXC chemokine in breast and kidney (BRAK), and MIP-2g, CXCL14 is a small cytokine in the CXC family. CXCL14 is expressed at high levels in many normal tissues, but is notably absent in many cancerous tissues. Further, CXCL14 plays a role in chemotaxis, homing, and activation for cells involved in the immune response and has been shown to inhibit angiogenesis.

Includes CXCL14 nucleic acid molecules and proteins. CXCL14 sequences are publicly available. For example, GenBank® Accession Nos. NM_004887.4, NM_001013137.2, and NM_019568.2 disclose exemplary human, rat, and mouse CXCL14 nucleotide sequences, respectively, and GenBank® Accession Nos. AAH03513.1, AAI01897.1, and AAH79661.1 disclose exemplary human, rat, and mouse CXCL14 protein sequences, respectively. One of ordinary skill in the art can identify additional CRISPS nucleic acid and protein sequences, including CXCL14 variants that retain CXCL14 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Differential expression or altered expression: A difference, such as an increase or decrease, in the conversion of the information encoded in a gene (such as a prostate cancer-related gene) into messenger RNA, the conversion of mRNA to a protein, or both. In some examples, the difference is relative to a control or reference value, such as a threshold value of expression for each marker, for example from one or more subjects (e.g., a cohort of control subjects). Detecting differential expression can include measuring a change in gene or protein expression, such as a change in expression of one or more prostate cancer-related genes or proteins disclosed herein.

Interleukin 24 (IL24): Also known as suppression of tumorigenicity 16 (ST16; e.g., OMIM 604136), melanoma differentiation-associated gene 7 (MDA7), ML-1, and IL-17F, IL24 is a cytokine and tumor-suppressing protein in the IL-10 family of cytokines. IL24 plays a role in cell survival and proliferation as well as wound healing, psoriasis, and cancer. Further, LI24 is expressed by cells involved in the immune response and can then act in skin, lung, and reproductive tissues.

Includes IL24 nucleic acid molecules and proteins. IL24 sequences are publicly available. For example, GenBank® Accession Nos. NM_006850.3, NM_133311.1, and NM_053095.2 disclose exemplary human, rat, and mouse IL24 nucleotide sequences, respectively, and GenBank® Accession Nos. NP_006841.1, NP_579845.1, and NP_444325.2 disclose exemplary human, rat, and mouse IL24 protein sequences, respectively. One of ordinary skill in the art can identify additional IL24 nucleic acid and protein sequences, including IL24 variants that retain IL24 biological activity (such as having increased levels in urine from a subject with prostate cancer).

Isolated: An “isolated” biological component (such as a nucleic acid molecule, protein, or cell) has been substantially separated or purified away from other biological components in the cell of the organism, or the organism itself, in which the component naturally occurs, such as other chromosomal and extra-chromosomal DNA and RNA, proteins and cells. Nucleic acid molecules and proteins that have been “isolated” include prostate cancer-related molecules (such as DNA or RNA) and proteins purified by standard purification methods. The term also embraces nucleic acid molecules, proteins and peptides prepared by recombinant expression in a host cell as well as chemically synthesized nucleic acid molecules and proteins. For example, an isolated protein, such as a prostate cancer-related protein, is one that is substantially separated from other types of proteins in a cell.

Label: An agent capable of detection, for example by mass spectrometry, ELISA, spectrophotometry, flow cytometry, or microscopy. For example, a label can be attached to a nucleic acid molecule or protein, thereby permitting detection of the nucleic acid molecule or protein. For example, a protein or peptide can be produced as a heavy, stable isotope, but as a protein or peptide with 13C or 15N incorporated as a heavy, stable isotope. Examples of labels include, but are not limited to, radioactive or heavy, stable isotopes, enzyme substrates, co-factors, ligands, chemiluminescent agents, fluorophores, haptens, enzymes, and combinations thereof. Methods for labeling and guidance in the choice of labels appropriate for various purposes are discussed for example in Sambrook et al. (Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, New York, 1989) and Ausubel et al. (In Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1998).

Matrix metalloproteinase 9 (MMP9): Also known as collagenase type IV B (CLG4B; e.g., OMIM 120361), gelatinase B (GELB), collagenase type V, and 92-KD gelatinase, MMP9 is a 92-kD type IV collagenase and a member of the zinc metalloproteinase family. MMP9 is expressed in cells involved in the immune response and has been found in skin, lung, and synovial tissues. Further, as a matrix metalloproteinase, MMP9 aids in breaking down the extracellular matrix for normal physiological processes; MMP9 is involved in the immune response, angiogenesis, and wound repair and is associated with autoimmune diseases, cancer, and cardiovascular disease.

Includes MMP9 nucleic acid molecules and proteins. MMP9 sequences are publicly available. For example, GenBank® Accession Nos. NM_004994.2, NM_031055.1, and NM_013599.4 disclose exemplary human, rat, and mouse MMP9 nucleotide sequences, respectively, and GenBank® Accession Nos. EAW75776.1, EDL96479.1, and EDL06438.1 disclose exemplary human, rat, and mouse MMP9 protein sequences, respectively. One of ordinary skill in the art can identify additional MMP9 nucleic acid and protein sequences, including MMP9 variants that retain MMP9 biological activity (such as being increased in the urine of prostate cancer patients, particularly intermediate- to high-risk prostate cancer).

Periostin (POSTN): Also known as PN (e.g., OMIM 608777) and osteoblast-specific factor 2 (OSF2), POSTN is an extracellular matrix protein. POSTN is expressed in many normal tissues, including aortis, stomach, lower gastrointestinal tract, placental, uterine, and breast tissues. Further, POSTN plays a role in tissue development and regeneration as well as epithelial cell adhesion and migration; POSTN is involved in cancer stem cell maintenance and metastasis.

Includes POSTN nucleic acid molecules and proteins. POSTN sequences are publicly available. Nucleic acid and protein sequences for POSTN are publicly available. For example, GenBank® Accession Nos. BC106709.1, NM_001108550.1, and BC031449.1 disclose exemplary human, rat, and mouse POSTN nucleotide sequences, respectively, and GenBank® Accession Nos. AAI06710.1, NP_001102020.1, and AAH31449.1 disclose exemplary human, rat, and mouse POSTN protein sequences, respectively. One of ordinary skill in the art can identify additional POSTN nucleic acid and protein sequences, including POSTN variants that retain POSTN biological activity (such as being increased in the urine of prostate cancer patients).

Prostate-specific antigen (PSA): Also known as kallikrein-related peptidase 3 (kallikrein 3, KLK3; e.g., OMIM 176820); antigen, prostate-specific (APS); and gamma-seminoprotein, PSA is a glycoprotein and member of the kallikrein-related peptidase family. PSA is predominantly secreted by epithelial cells in the prostate gland and functions to dissolve cervical mucus to facilitate sperm entry into the uterus. PSA has been used to diagnose prostate cancer, as increased PSA levels in blood may suggest the presence of prostate cancer.

Includes PSA nucleic acid molecules and proteins. PSA sequences are publicly available. Nucleic acid and protein sequences for PSA are publicly available. For example, GenBank® Accession Nos. NM_001648.2, NM_012725.2, and NM_008455.3 discloses exemplary human PSA nucleotide sequences, respectively, and GenBank® Accession Nos. CAD54617.1, AAH89815.1, and NP_001639.1 discloses exemplary human PSA protein sequences. One of ordinary skill in the art can identify additional PSA nucleic acid and protein sequences, including PSA variants that retain PSA biological activity (such as being secreted by the prostate gland).

Prostate cancer: Also known as carcinoma of the prostate, prostate cancer is the development of cancer in the prostate, a gland in the male reproductive system. Prostate cancer is classified into different risk categories, including low-, intermediate-, and high-risk prostate cancer, which means that a patient has a low-, intermediate-, and high-risk, respectively, of pathological and biochemical outcomes after radical prostatectomy; metastasis; prostate cancer-specific mortality; and all-cause mortality (Cooperberg et al., J Cancer Inst., 101(12):878-887, 2009). One means of assessing the risk is using Gleason scoring: low-risk prostate cancer, Gleason score sum less than or equal to 6; intermediate-risk prostate cancer, Gleason score sum at 7; and high-risk prostate cancer, Gleason score sum greater than 7. Most prostate cancers are slow growing; however, some grow relatively quickly. The cancer cells may spread from the prostate to other parts of the body, particularly the bones and lymph nodes. It may initially cause no symptoms. In later stages, it can lead to difficulty urinating, blood in the urine, or pain in the pelvis, back or when urinating or to feeling tired due to low levels of red blood cells.

Prostate cancer can be diagnosed by biopsy. Medical imaging may then be done to determine if the cancer has spread to other parts of the body. Prostate cancer screening is controversial. Prostate-specific antigen (PSA) testing increases cancer detection but does not decrease mortality. The United States Preventive Services Task Force recommends against screening using the PSA test, due to the risk of overdiagnosis and overtreatment, as most cancer diagnosed would remain asymptomatic, and concludes that the potential benefits of testing do not outweigh the expected harms.

Many cases can be safely followed with active surveillance or watchful waiting. Other treatments may include a combination of surgery (such as cryotherapy), radiation therapy, hormone therapy, and chemotherapy. When it only occurs inside the prostate it may be curable. In those in whom the disease has spread to the bones, pain medications, bisphosphonates and targeted therapy, among others, may be useful. Outcomes depend on a person's age and other health problems as well as how aggressive and extensive the cancer is. Most people with prostate cancer do not die from the disease. The 5-year survival rate in the United States is 99%. Globally, it is the second most common type of cancer and the fifth leading cause of cancer-related death in men. Studies of males who died from unrelated causes have found prostate cancer in 30% to 70% of those over age 60.

Sample: A biological specimen containing genomic DNA, RNA (e.g., mRNA), protein, or combinations thereof, obtained from a subject. Examples include, but are not limited to, peripheral blood, serum, plasma, urine, ejaculate, saliva, tissue biopsy, fine needle aspirate, surgical specimen, and autopsy material. In one example, a sample is a urine sample from a subject with or at risk for prostate cancer, such as low-, intermediate-, or high-risk prostate cancer. In some examples, samples are used directly in the methods provided herein. In some examples, samples are manipulated prior to analysis using the disclosed methods, such as through concentrating, filtering, centrifuging, diluting, desalting, denaturing, reducing, alkylating, proteolyzing, or combinations thereof. In some examples, components of the samples are isolated or purified prior to analysis using the disclosed methods, such as isolating cells, proteins, and/or nucleic acid molecules from the samples.

Secreted frizzled-related protein 4 (SFRP4): Also known as frizzled-related protein (e.g., OMIM 606570) and human endometrium (FRPHE), SFRP4 is in the SFRP family, the members of which regulate Wnt signaling. SFRP4 is expressed in the endometrium, myocardium, breast tissue, and islets. Further, SFRP4 plays a role in regulating apoptosis, insulin secretion, and in regulating uterine morphology and function. SFRP4 is associated with bone diseases, such as rickets and bone cancers.

Includes SFRP4 nucleic acid molecules and proteins. SFRP4 sequences are publicly available. For example, GenBank® Accession Nos. NM_003014.3, NM_053544.1, and NM_016687.3 disclose exemplary human, rat, and mouse SFRP4 nucleotide sequences, respectively, and GenBank® Accession Nos. CAG46532.1, NP_445996.1, and AAH34853.1 disclose exemplary human, rat, and mouse SFRP4 protein sequences, respectively. One of ordinary skill in the art can identify additional SFRP4 nucleic acid and protein sequences, including SFRP4 variants that retain SFRP4 biological activity (such as being increased in the urine of prostate cancer patients).

Subject: Living multi-cellular vertebrate organisms, a category that includes mammals, such as human and non-human mammals, such as veterinary subjects (for example cats, dogs, cows, sheep, horses, pigs, and mice). In a particular example, a subject is one who has or is at risk for prostate cancer, such as low-, intermediate-, or high-risk prostate cancer. In a particular example, a subject is one who is suspected of having prostate cancer.

Therapeutically effective amount: An amount of a pharmaceutical preparation that alone, or together with a pharmaceutically acceptable carrier or one or more additional therapeutic agents, induces the desired response. A therapeutic agent, such as an anti-neoplastic chemotherapeutic agent, radiotherapeutic agent, or biologic agent, is administered in therapeutically effective amounts.

Therapeutic agents can be administered in a single dose, or in several doses, for example daily, during a course of treatment. However, the effective amount can be dependent on the source applied, the subject being treated, the severity and type of the condition being treated, and the manner of administration. Effective amounts of a therapeutic agent can be determined in many different ways, such as assaying for a sign or a symptom of an adenocarcinoma. Effective amounts also can be determined through various in vitro, in vivo or in situ assays. For example, a pharmaceutical preparation can decrease one or more symptoms of a prostate cancer, for example, a decrease in the size of the prostate cancer, the number of tumors, the number of metastases, or other symptoms (or combinations thereof) by at least 20%, at least 50%, at least 70%, at least 90%, at least 98%, or even 100%, as compared to an amount in the absence of the pharmaceutical preparation.

Treating a disease: “Treatment” refers to a therapeutic intervention that ameliorates a sign or symptom of a disease or pathological condition, such a sign or symptom of prostate cancer. Treatment can also induce remission or cure of a condition, or can reduce the pathological condition, such as a reduction in tumor size, a reduction in tumor burden, a reduction in a sign or a symptom of a tumor (such as cachexia), a reduction in metastasis, or combinations thereof. In particular examples, treatment includes preventing a disease, for example by inhibiting the full development of a disease, such as decreasing the ability of a tumor to metastasize. Prevention of a disease does not require a total absence of disease.

Upregulated or activation: When used in reference to the expression of a nucleic acid molecule, such as a gene, refers to any process which results in an increase in the production of a gene product. A gene product can be RNA (such as mRNA, rRNA, tRNA, and structural RNA) or protein. Therefore, gene upregulation or activation includes processes that increase transcription of a gene or translation of mRNA, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4.

Examples of processes that increase transcription include those that facilitate formation of a transcription initiation complex, those that increase transcription initiation rate, those that increase transcription elongation rate, those that increase processivity of transcription, and those that relieve transcriptional repression (for example, by blocking the binding of a transcriptional repressor). Gene upregulation can include inhibition of repression as well as stimulation of expression above an existing level. Examples of processes that increase translation include those that increase translational initiation, those that increase translational elongation and those that increase mRNA stability.

Gene upregulation includes any detectable increase in the production of a gene product, such as a protein. In certain examples, production of a gene product increases by at least 2-fold, at least 3-fold, at least 4-fold, or at least 5-fold, as compared to a control (e.g., as compared to a threshold of expression of any of these molecules established from a subject or subjects, such as a cohort of control subjects).

Overview

Provided herein are prostate cancer-associated secreted proteins that are found in increased levels in the urine of patients with prostate cancer, as compared to levels in urine from patients without prostate cancer. Sensitive multiplexed assays were developed for reliable simultaneous quantification of 12 detectable secreted proteins in urine. Except for CCL3 and POSTN, the other 10 proteins were reproducibly detected and quantified in all the urine samples analyzed with at least one surrogate peptide. The peptide signatures from AGR2, AGR3, CEACAM5, CD90 and SFRP4, in particular, produced an area-under-the curve (AUC) of 0.95. It was also observed that urinary MMP9 levels increased with higher risk cancer, which correlated with increase in MMP9 gene expression in Gleason pattern≥7 vs. Gleason≤6 prostate cancer subjects.

Samples of voided urine (≤100 ml) were collected without DRE from non-cancer controls, pre-op(erative), and post-op patients; post-pellet urine supernatant was spin concentrated (to <1 ml); urinary proteins were simultaneously measured by sensitive multiplex targeted proteomics. The quantification accuracy of each surrogate peptide assay was evaluated by correlation analysis of relative protein abundance between surrogate peptides from the same protein. The ability of measured markers to distinguish cancer from non-cancer was assessed by AUC values.

The secreted protein cancer-related biomarkers disclosed herein can generate an AUC over 0.9 for detecting prostate cancer. These markers were identified from their elevated expression in cell types of prostate tumors. Multimarker measurement can be accurately obtained, for example by using proteomics tools.

Accuracy in SRM assays depends on the surrogate peptides selected for each biomarker; the measured concentrations are directly proportional to the cognate protein analytes. As shown by the PSA surrogate peptides examined herein, unknown modifications on the analyte peptide sequences produce lower levels of measured concentrations compared with the true concentrations. Without modifications, the abundance ratio for any two or more surrogate peptides is constant across multiple samples analyzed, and a good correlation curve with no significant data point deviation is produced. Based on the correlation coefficients, the quantification accuracy of individual selected surrogate peptides of the secreted protein markers was assured, and the surrogate peptides were selected for SRM assay configuration for all subsequent testing. In studies involving human cell lines, most surrogate peptides (453/466) generated a high correlation coefficient (R2>0.8) with no significant data point deviation (Worboys et al., Nat Methods 11:1041-4, 2014). Many surrogate peptides used in the urine analysis generated lower correlation coefficients (the median R2=0.70) with data point deviations. This shows that the targeted protein molecules in urine of multiple patient samples are more varied, such as due to allelic differences or cancer-associated isoforms, than those in single cell lines. PSA measurement in urine, for example, was shown to be dependent on the peptide chosen. A single PSA peptide could not accurately quantify PSA in three of the urine samples (FIG. 8, urine IDs. P06017Pre, P07040Pre, and P08015Pre). A specific PSA proteoform encoded by SNP-L132I (rs2003783) is located within the LSEPAE(L/I)TDAVK (SEQ ID NO: 71) surrogate peptide. This newly identified PSA proteoform was observed in 9 out of 72 clinical serum samples (Végvári et al., Mol Cell Proteomics 12:2761-73, 2013) and in the P06017Pre urine. Abundance recalculations based on this information improved the correlation for this urine sample.

In addition to surrogate peptide selection, PSA normalization was used to account for the contribution of protein markers from sources other than the prostate. Without PSA normalization, the median values of the marker protein L/H ratios were lower in the non-cancer urine than the cancer urine, while, for CRISP3 and SFRP4, the median values in the non-cancer urine were higher than the cancer urine. For example, the L/H ratio for WANQCNYR (SEQ ID NO: 12) from CRISP3 in cancer was 0.29, and 0.38 in non-cancer. Given that CRISP3 and SFRP4 are produced by other sources in the urinary tract, this confounding result was not unexpected. Expression of CRISP3 and the non-secreted prostate cancer marker AMACR is localized to other tissues in the urinary tract. A similar finding for AGR2 in urine was obtained, even though a database query showed no detectable AGR2 in normal urine. One set of urine samples produced a 0.002 L/H ratio in cancer compared to 0.005 in non-cancer (Shi et al., J Proteome Res 13:875-82, 2014). AGR2 can originate from several other sources, such as the bladder and kidney. Although normal AGR2-expressing bladder urothelial cells do not secrete AGR2, unknown physiological factors could produce detectable levels of AGR2 in urine. While urinary CRISP3 and SFRP4 are constitutively produced by non-prostate sources, urinary AGR2 is transiently produced from non-prostate sources. Urinary PSA, on the other hand, is exclusively produced by the prostate. The significantly higher concentrations of urinary PSA found in some non-cancer samples could be due to donors with an enlarged prostate from benign hyperplasia. For example, prostate cancer patients with a prostate volume of 35 cm3 (n=29) and benign prostatic hyperplasia patients prostate volume of 45 cm3 (n=35) were measured to have median urinary PSA levels of 52.6 ng/mL and 123.2 ng/mL, respectively (Bolduc et al., Can Urol Assoc J 1:377-81, 2007). With PSA normalization, the performance of the disclosed prostate cancer protein biomarkers showed a near perfect AUC.

The use of the disclosed prostate cancer-related biomarkers reduces the need for prostate biopsy. In one example, prostate cancer diagnosis includes the use of the disclosed prostate cancer-related markers before or following an abnormal serum PSA result. Urine donation is convenient and does not require a DRE. With regard to the possibility that DRE might enhance marker signals, the levels of two post-DRE urine, P08-032C and P08-036N, were obtained. The signals from P08-032C (Gleason 4+3, tumor volume 1 cc) were not higher than those from others obtained without a DRE. DRE did not produce any increased signals in non-cancer P08-036N. With regard to possible age-related increases in marker levels, samples from a 76-year-old man (P08-018N) and a 53-year-old man (P08-022N) were analyzed. There was no detected increase in the background urinary levels of these proteins. It is possible that the baseline level of these markers may remain more or less constant with age. In contrast, an increase in serum PSA with age is known. In one example, if the prostate cancer-related protein panel result is negative (e.g., the analyzed proteins are not increased in the urine), no biopsy would be necessary if the negative predictive value is sufficiently high. Furthermore, the ratio of MMP9/PSA concentrations can be used to distinguish low volume/low grade prostate cancer from significant cancer. Therefore, effectively integrating the results from analyzing expression of the disclosed prostate cancer-related molecules will lead to greater detection of significant cancer with fewer biopsies performed in patients without cancer.

For AGR2 quantification, both mass spectrometry proteomics and ELISAs were used with good correlation, R2=0.91, and similar AUC values (0.75). Thus, the disclosed prostate cancer-related biomarkers can be used for a multiplex ELISA to measure all these proteins simultaneously in a clinical setting. The equivalency in urinary AGR2 quantification by PRISM-SRM and ELISA shows that a multiplex ELISA can replicate the proteomics results, and clinical proteomics can make large-scale testing feasible.

Evaluating Expression in a Subject with a Risk of Prostate Cancer

Provided herein are methods of diagnosing a subject with a risk of prostate cancer and methods of treating a subject with prostate cancer (such as a human or veterinary subject). In particular examples, the methods can determine with high specificity, sensitivity, and accuracy (such as having an AUC of greater than 0.8, including, for example, at least 0.85, at least 0.9, at least 0.95, at least 0.96, at least 0.97, at least 0.98, or at least 0.99) whether a subject is likely to have prostate cancer. The prostate cancer can be any risk category of interest, including low- (Gleason score sum is 6 or lower), intermediate- (Gleason score sum is 7), and high-risk (Gleason score sum is above 7) prostate cancer. It is helpful to be able to determine whether or not a subject has prostate cancer because there are a variety of protocols for diagnosing prostate cancer but not all are specific, sensitive, and accurate. Hence, using the results of the disclosed assays to help distinguish subjects that are likely to have prostate cancer versus those not likely to have prostate cancer offers a substantial clinical benefit and allows subjects to be accurately diagnosed and, if a subject has prostate cancer, to be accurately treated.

In additional examples, the methods are utilized to determine whether or not to provide the subject with therapeutic intervention. In one example, a therapeutic intervention is administered. Thus, if the subject has prostate cancer, a therapeutic intervention, such as watchful waiting, active surveillance, surgery, radiation, hormone therapy, chemotherapy, brachytherapy, cryotherapy, ultrasound, bisphosphate therapy, biologic therapy, or vaccine therapy can be utilized. Using the results of the disclosed assays to help distinguish subjects that are likely to have prostate cancer versus those not likely to have prostate cancer offers a substantial clinical benefit because, where the subject has prostate cancer, the methods disclosed herein allow the subject to be selected for therapeutic intervention.

Methods of diagnosing a subject with a risk of prostate cancer and methods of treating a subject with prostate cancer, such as low-, intermediate-, or high-risk prostate cancer, are provided. Such methods can include measuring or detecting absolute or relative amounts of prostate cancer-related markers present in a sample (such as a urine sample) obtained from the subject, for example, using surrogate peptides of the marker proteins (e.g., as shown in FIGS. 4, 12 and 15) and/or antibodies, nucleic acid probes, and/or nucleic acid primers specific for each marker. In some examples, the prostate cancer-related markers can include at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11 or all 12 of (such as 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 of) AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4. The expression levels of these markers can be measured. If increased protein and/or nucleic acid expression of the prostate cancer-related markers, for example, expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, in the sample is measured, the method can include administering therapeutic intervention to the subject, thereby treating the subject.

In some examples, measuring expression of prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, can include quantitating protein and/or nucleic acid expression of these markers in a sample obtained from the subject. In particular examples, these markers are first analyzed for measurement accuracy, such as correlating the amounts of different surrogate peptides from the same prostate cancer-related marker protein where the protein expression is measured.

In other examples, measuring increased protein or nucleic acid expression of the markers AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 is relative to an amount of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 median protein or nucleic acid expression, respectively, for example a median value of protein or nucleic acid expression for each marker expected in a subject with no prostate cancer (e.g., as compared to a threshold of expression of any of these molecules established from a subject or subjects, such as a cohort of control subjects).

In some examples, measuring protein and/or nucleic acid expression of the markers AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 can include measuring more than one marker, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or all 12 of the markers. In other examples, any combination of these markers can be measured. In particular examples, any of the combinations of markers listed in FIG. 15 can be measured.

In some examples, measuring expression of prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, can include measuring the amount of protein expressed. For example, measuring the amount of protein expressed can include measuring a surrogate peptide from the protein. More than one surrogate peptide can be measured for a marker (e.g., see FIG. 2, FIG. 4, FIGS. 11A-B, FIGS. 13A-B, and FIGS. 18A-B), such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, or at least 20 (such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 20) surrogate peptides for a single marker. In some specific examples, surrogate peptides can be generated through contacting the protein (such as a sample containing the protein) with a protease, such a trypsin. Thus, in some examples, the methods includes treating a sample to be analyzed with a protease, such as trypsin. In some particular examples, surrogate peptides for the prostate cancer-related markers AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 include the surrogate peptides listed in FIG. 4. In other particular examples, combinations of surrogate peptides for prostate cancer-related markers can be used, such as the combinations listed in FIG. 15.

In some examples, measuring the amount of protein expressed for prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, can include using mass spectroscopy and/or an immunoassay.

In particular examples, measuring the amount of protein expressed can include measuring the protein concentration using an immunoassay, such as an ELISA.

In some examples, measuring expression of prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, using the amount of protein expressed can include measuring the protein concentration using mass spectrometry. In some examples, mass spectrometry can be used to determine the protein concentration of the full-length protein and/or surrogate peptide(s) for the protein. In particular examples, mass spectrometry can be used to determine the protein concentration of prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, using surrogate peptides, such as the peptides listed in FIG. 4.

In particular examples, measuring expression of prostate cancer-related markers by using mass spectrometry can include using mass spectrometry assays such as LC-SRM, LG-SRM, and/or PRISM-SRM. In some examples, measuring expression of prostate cancer-related markers (such as in a serum sample) can include using an LC-SRM assay, for example, where the serum protein levels are least at a moderate abundance, such as about low μg/mL (e.g., 1-10, 10-50, 50-100, or 100-500 μg/mL).

In other examples, the measuring increased protein or nucleic acid expression of the markers AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 includes measuring some markers that are at low-to-moderate abundance, for example, in the range of about low μg/mL to high ng/mL (e.g., 1-10 μg/mL, 500 ng/mL-1 μg/mL, or 100-500 ng/mL), in the sample obtained from the subject. In particular examples, the low-to-moderate-abundance markers in the sample can include CRISP3, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4. In some examples, these low-abundance markers can be accurately measured by using assays with sufficient sensitivity, such as an LG-SRM assay, a PRISM-SRM assay, and/or an ELISA.

In certain examples, measuring increased protein or nucleic acid expression of the markers AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 includes measuring some markers that are at low abundance, for example, in the range of about low ng/mL to high pg/mL (e.g., 500-100 ng/mL, 100-50 ng/mL, 50-10 ng/mL, 10-1 ng/mL, 500 pg/mL-1 ng/mL, 500-100 pg/mL, or 100-50 pg/mL) in the sample obtained from the subject. In particular examples, the low-abundance markers in the sample can include AGR2, AGR3, CCL3, CEACAM5, and CEACAM6. In some examples, these low-abundance markers can be accurately measured by using assays with sufficient sensitivity, such as a PRISM-SRM assay and/or an ELISA.

In other examples, measuring increased protein or nucleic acid expression of prostate cancer-related markers, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, includes normalizing expression of the prostate cancer-related molecules (such as surrogate peptides provided herein, such as those shown in FIG. 4 or 15) to the expression of a prostate molecule. In particular examples, the prostate molecule used for normalization can be PSA. In specific examples, normalizing to the PSA concentration can include normalizing the protein expression of a prostate cancer-related marker to the amount of PSA protein. In other examples, normalizing to the amount of PSA can include normalizing the protein expression of a prostate cancer-related marker to the amount of at least one surrogate peptide from the PSA protein. In certain examples, the PSA protein surrogate peptide(s) can include IVGGWECEK, LSEPAELTDAVK, or both.

In some examples, the methods can include measuring increased expression of two or more prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, which can include the combinations of markers listed in FIG. 15. In particular examples, the protein expression of the markers can be measured, for example, by using surrogate peptides of the markers, such as the surrogate peptides listed in FIG. 4. More than one surrogate peptide can be used for a marker (e.g., FIG. 2, FIG. 4, FIGS. 11A-B, FIGS. 13A-B, and FIGS. 18A-B), such as at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, or at least 20 (such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or 20) surrogate peptides for a single protein marker. In specific examples, the amounts of surrogate peptides can be measured using mass spectrometry, such as LC-SRM, LG-SRM, and/or PRISM-SRM. In another example, the amounts of surrogate peptides are normalized to a prostate molecule, such as PSA, for example, by using one or more surrogate peptides of PSA (e.g., IVGGWECEK, SEQ ID NO: 70, LSEPAELTDAVK, SEQ ID NO: 71, or both). In specific examples, the increased expression measured for the at least two prostate cancer-related markers (such as 3, 4 5, 6, 7, 8, 9, 10, 11 or all 12 of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4) has an AUC value greater than 0.80, such as an AUC value of 0.81, 0.82, 0.83, 0.84, 0.85, 0.86, 0.87, 0.88, or 0.89. In other specific examples, the increased expression measured for the at least two prostate cancer-related markers (such as 3, 4 5, 6, 7, 8, 9, 10, 11 or all 12 of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4) has an AUC value greater than 0.90, such as an AUC value of 0.91, 0.92, 0.93, 0.94, 0.95, 0.96, 0.97, 0.98, or 0.99.

Methods of diagnosing intermediate- or high-risk prostate cancer and treating a subject with intermediate- or high-risk prostate cancer are provided. In particular examples, the methods can determine with significant accuracy whether a subject is likely to have prostate cancer associated with a specific risk category, such as low-, intermediate-, and high-risk prostate cancer. In particular examples, the methods can distinguish with significant accuracy between subjects that have low-risk prostate cancer and subjects that have intermediate-to-high-risk prostate cancer. It is helpful to be able to determine whether or not a subject has intermediate-to-high-risk prostate cancer because there are a variety of protocols for treating prostate cancer. Hence, using the results of the disclosed assays to help distinguish subjects that are likely to have prostate cancer associated with a specific risk category offers a substantial clinical benefit and allows subjects to be accurately diagnosed and, if a subject has prostate cancer associated with a specific risk category, to be accurately treated.

In some examples, methods of diagnosing intermediate- or high-risk prostate cancer and treating a subject with intermediate- or high-risk prostate cancer can include measuring a sample (such as a urine sample) obtained from the subject for example, using surrogate peptides of the marker proteins and/or antibodies, nucleic acid probes, and/or nucleic acid primers specific for the markers. In some examples, the prostate cancer-related markers can include MMP9. The expression levels of the markers can be measured. If increased protein and/or nucleic acid expression of one or more prostate cancer-related markers, for example, expression of MMP9, in the sample is measured, the method can include administering therapeutic intervention for intermediate- or high-risk prostate cancer to the subject, thereby treating the subject.

In particular examples, expression of MMP9 in a sample obtained from a subject is measured. In one example, the expression of MMP9 can be determined based on MMP9 protein concentration. In some examples, the expression of MMP9 protein can be measured using at least one surrogate peptide of MMP9. In specific examples, the surrogate peptide can be FQTFEGDLK (SEQ ID NO: 41), LGLGADVAQVTGALR (SEQ ID NO: 42), or both.

In further examples, measuring increased expression of MMP9 in the sample obtained from the subject can include comparing the expression of MMP9, such as by using the concentration of the MMP9 surrogate peptide(s) FQTFEGDLK (SEQ ID NO: 41) and/or LGLGADVAQVTGALR (SEQ ID NO: 42), to the amounts of MMP9 expression expected in a sample from a subject who has low-risk prostate cancer. In some specific examples, the surrogate peptides FQTFEGDLK (SEQ ID NO: 41) and LGLGADVAQVTGALR (SEQ ID NO: 42) for MMP9 protein can be used to measure expression of MMP9 with significant accuracy. In certain examples, the expression of MMP9 is increased compared to a sample from one or more subjects (such as a cohort of control subjects) who has low-risk prostate cancer, such as where the expression of MMP9 is increased compared with a sample from a subject who has low-risk prostate cancer with a P value of less than 0.05, such as a P value of 0.022 or a range of P values between 0.01-0.02, 0.02-0.03, 0.03-0.04, or 0.05.

In some examples, where an increase in the expression of MMP9 is measured in the sample obtained from the subject compared with the expression of MMP9 expected in a sample from a subject who has low-risk prostate cancer, the methods include administering treatment for intermediate- or high-risk prostate cancer, thereby treating the subject. For example, a patient with low-risk prostate cancer may not exhibit increased expression of MMP9 that exceeds the expression expected from a patient with low-risk prostate cancer and, therefore, may not necessarily be a good candidate for invasive treatments and/or treatments with potentially harmful side effects but, rather, may be a good candidate for watchful waiting or active surveillance. In another example, a patient with intermediate-to-high-risk prostate cancer may exhibit increased levels of MMP9 compared with the expression expected from a patient with low-risk prostate cancer and, therefore, may be better candidate for treatments such as surgery, radiation therapy, and hormone therapy but may not be a good candidate for observation-based treatments, such as watchful waiting or active surveillance.

Evaluating Nucleic Acid Expression

In some examples, expression of nucleic acids (e.g., RNA, mRNA, cDNA, genomic DNA) of prostate cancer-related markers, such as the markers AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, are analyzed and, in some examples, quantified. Suitable biological samples can include urine, blood, plasma, or serum samples obtained from a subject having or a subject at risk for prostate cancer (such as intermediate- or high-risk prostate cancer). An increase in the amount of nucleic acid molecules for the prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, in the sample indicates that the subject has prostate cancer and/or has intermediate- or high-risk prostate cancer, as described herein. In some examples, expression of the prostate cancer-related nucleic acid molecule is normalized to PSA expression in the sample (such as by measuring PSA cDNA, genomic DNA, or mRNA in the urine sample). In some examples, the assay is multiplexed, in that expression of several nucleic acids are detected simultaneously or contemporaneously (Quek et al., Prostate 75:1886-95, 2015).

Nucleic acid molecules can be isolated from a sample from a subject having or a subject at risk for prostate cancer or for intermediate- or high-risk prostate cancer, such as a urine, blood, plasma, or serum sample. In one example, RNA isolation is performed using a purification kit, buffer set, and protease from commercial manufacturers, such as QIAGEN®, according to the manufacturer's instructions. RNA prepared from a biological sample can be isolated, for example, by guanidinium thiocyanate-phenol-chloroform extraction, and oligp(dT)-cellulose chromatography (e.g., Tan et al., J Biomed Biotechnol., 2009: 574398, 10 pages, incorporated herein by reference in its entirety).

Methods of gene expression profiling include methods based on hybridization analysis of polynucleotides, methods based on sequencing of polynucleotides, and other methods in the art. In some examples, mRNA expression is quantified using northern blotting or in situ hybridization; RNAse protection assays, or PCR-based methods, such as reverse transcription polymerase chain reaction (RT-PCR) or real time quantitative RT-PCR. Alternatively, antibodies can be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. Representative methods for sequencing-based gene expression analysis include Serial Analysis of Gene Expression (SAGE) and gene expression analysis by massively parallel signature sequencing (MPSS).

Evaluating Protein Expression

In some examples, protein expression of prostate cancer-related markers, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, is analyzed and, in some examples, quantified. Suitable biological samples include urine, ejaculate, blood, plasma, and/or serum samples obtained from a subject having or a subject at risk for prostate cancer, such as for intermediate- or high-risk prostate cancer. An increase in the amount of prostate cancer-related marker proteins, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins, in the sample indicates that the subject has prostate cancer and/or has intermediate- or high-risk prostate cancer, as described herein. In some examples, expression of the prostate cancer-related protein is normalized to PSA expression in the sample (such as by measuring a surrogate peptide(s) for PSA in the urine sample). In some examples, the assay is multiplexed, in that expression of several proteins is detected simultaneously or contemporaneously.

The expression of prostate cancer-related markers, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4, can be measured using any of a number of techniques, such as direct physical measurements (e.g., mass spectrometry) or binding assays (e.g., immunoassays, agglutination assays, and immunochromatographic assays, such as ELISA, Western blot, or RIA assay). Immunohistochemical techniques can also be utilized for protein detection and quantification.

The method can include measuring or detecting a signal that results from a chemical reaction, e.g., a change in optical absorbance, a change in fluorescence, the generation of chemiluminescence or electrochemiluminescence, a change in reflectivity, refractive index or light scattering, the accumulation or release of detectable labels from the surface, the oxidation or reduction or redox species, an electrical current or potential, changes in magnetic fields, etc. Suitable detection techniques can detect binding events by measuring the participation of labeled binding reagents through the measurement of the labels via their photoluminescence (e.g., via measurement of fluorescence, time-resolved fluorescence, evanescent wave fluorescence, up-converting phosphors, multi-photon fluorescence, etc.), chemiluminescence, electrochemiluminescence, light scattering, optical absorbance, radioactivity, magnetic fields, enzymatic activity (e.g., by measuring enzyme activity through enzymatic reactions that cause changes in optical absorbance or fluorescence or cause the emission of chemiluminescence). In some examples, detection techniques are used that do not require the use of labels, e.g., techniques based on measuring mass (e.g., surface acoustic wave measurements), refractive index (e.g., surface plasmon resonance measurements), or the inherent luminescence of an analyte, such as a prostate cancer-related marker, for example, AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4.

For the purposes of quantitating proteins, a biological sample of the subject that includes cellular proteins (such as urine) can be used. Quantitation of prostate cancer-related marker proteins, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins, can be achieved by immunoassay. The amount of prostate cancer-related marker proteins, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins, can be assessed in the sample, for example by contacting the sample with appropriate antibodies (or antibody fragments) specific for each protein, and then detecting a signal (for example present directly or indirectly on the antibody, for example by the use of a labeled secondary antibody).

In one example, an electrochemiluminescence immunoassay is used, such as the V-PLEX™ system (Meso Scale Diagnostics, Rockville, Md.). In such assays, the primary antibodies for prostate cancer-related marker proteins, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins, (or the corresponding secondary antibodies) are labeled with an electrochemiluminescent label.

Quantitative spectroscopic approaches methods, such as LC-SRM, LG-SRM, PRISM-SRM, and surface-enhanced laser desorption-ionization (SELDI), can be used to analyze expression of prostate cancer-related marker proteins, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins, in, for example, a urine sample obtained from a subject having or a subject at risk for prostate cancer, such as for intermediate- or high-risk prostate cancer. In some such spectroscopy methods, at least one surrogate peptide for each prostate cancer-related marker protein is measured or detected in the sample (e.g., see FIGS. 4 and 15).

In one example, LC-SRM (liquid chromatography-selected reaction monitoring) may be used to detect protein expression for example by using a triple quadrupole spectrometer (see, e.g., U.S. Pub. No. 2013/0203096). LC-SRM is a liquid chromatography method that can be used for high-throughput selective and sensitive detection of molecules, such as prostate cancer-related proteins, for example, AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4. It can quantify moderately abundant analytes (low μg/mL) in limited sample volumes.

Therefore, in a particular example, the analytes can include prostate cancer-related marker proteins and/or surrogate peptides thereof, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins and/or surrogate peptides thereof. In other examples, the fractionated and pooled analytes consist essentially of or consist of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 proteins or surrogate peptides thereof or of the combinations of proteins or surrogate peptides listed in FIG. 15. In this context, “consists essentially of” indicates that the fractionated and pooled analytes do not include other prostate cancer-related marker proteins that can be used to accurately predict prostate cancer, but can include other prostate molecules, such as prostate protein expression controls (e.g., PSA protein or surrogate peptides thereof).

In another example, LG-SRM (long gradient-selected reaction monitoring) can be used to detect protein expression, for example by using a reversed-phase C18 column and triple quadrupole spectrometer (see, e.g., Shi et al., Anal Chem., 85(19):9196-9203). LG-SRM is a liquid chromatography method for sensitive quantitation of analytes, such as prostate cancer-related proteins, and can even be used to accurately quantitate low-to-moderately abundant analytes (low μg/mL to high ng/mL), such as CRISP3, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4.

In LG-SRM, a long, shallow LC gradient (e.g., 5 hours compared with a conventional LC protocol that can be about 45 min) using a long LC column is followed by SRM as a second step. The eluting LC peaks containing the target analyte, such as prostate cancer-related proteins or surrogate peptides thereof, for example, AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins or surrogate peptides thereof, are, thus, sufficiently separated and resolved for accurate quantitation via SRM.

Therefore, in a particular example, the target analytes include prostate cancer-related marker proteins and/or surrogate peptides thereof, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins and/or surrogate peptides thereof. In other examples, the target analytes consist essentially of or consist of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 proteins or surrogate peptides thereof; of the combinations of proteins or surrogate peptides listed in FIG. 15; or of moderate-to-low-abundance proteins or surrogate peptides thereof, such as CRISP3, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4. In this context “consists essentially of” indicates that the target analytes do not include other prostate cancer-related marker proteins that can be used to accurately predict prostate cancer, but can include other prostate molecules, such as prostate protein expression controls (e.g., PSA protein or surrogate peptides thereof).

In an additional example, PRISM-SRM (high-pressure, high-resolution separations, intelligent selection, multiplexing-selected reaction monitoring) is used to detect protein expression, for example, by using an ultra-pressure LC (UPLC) system and a triple quadrupole spectrometer (see, e.g., U.S. Pub. No. 2014/0194304; Shi et al., PNAS, 109(38):15395-15400 (2012); and Shi et al., J Proteome Res., 13(2):875-882 (2014)). PRISM-SRM is a liquid chromatography method for quantitating analyes, such as prostate cancer-related proteins, and can even be used to accurately quantitate low-abundance (low ng/mL to high pg/mL) analytes, such as AGR2, AGR3, CCL3, CEACAM5, and CEACAM6.

In PRISM-SRM, LC-SRM is used as a second step after the target analyte is enriched through a liquid chromatography pre-fractionation step, such as using reverse-phase chromatography. The fractions containing the target analyte, such as prostate cancer-related proteins or surrogate peptides thereof, for example, AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins or surrogate peptides thereof, can then be pooled. The pooled fractions are enriched in the target analyte(s) and can then undergo a second LC separation step followed by SRM analysis.

Therefore, in a particular example, the fractionated and pooled analytes include prostate cancer-related marker proteins and/or surrogate peptides thereof, such as AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and/or SFRP4 proteins and/or surrogate peptides thereof. In other examples, the fractionated and pooled analytes consist essentially of or consist of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 proteins or surrogate peptides thereof; of the combinations of proteins or surrogate peptides listed in FIG. 15; or of low-abundance proteins or surrogate peptides thereof, such as AGR2, AGR3, CCL3, CEACAM5, and CEACAM6. In this context “consists essentially of” indicates that the fractionated and pooled analytes do not include other prostate cancer-related marker proteins that can be used to accurately predict prostate cancer, but can include other prostate molecules, such as prostate protein expression controls (e.g., PSA protein or surrogate peptides thereof).

In a further example, surface-enhanced laser desorption-ionization time-of-flight (SELDI-TOF) mass spectrometry is used to detect protein expression, for example by using the ProteinChip™ (Ciphergen Biosystems, Palo Alto, Calif.).

Prostate Cancer-Related Molecules

The disclosed prostate cancer-related molecules include AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, SFRP4, and PSA. One or more of the disclosed prostate cancer-related molecules can be used alone or in any combination. The molecules can include proteins, peptides (e.g., surrogate peptides, see FIGS. 2, 4, and 15 for example), and nucleic acids.

In some embodiments, one of the disclosed prostate cancer-related molecules can include AGR2 (e.g., SEQ ID NOS: 1-3). In some embodiments, one of the disclosed prostate cancer-related molecules can include AGR3 (e.g., SEQ ID NOS: 4-9). In some embodiments, one of the disclosed prostate cancer-related molecules can include CRISP3 (e.g., SEQ ID NOS: 10-15). In some embodiments, one of the disclosed prostate cancer-related molecules can include CCL3 (e.g., SEQ ID NOS: 16-20). In some embodiments, one of the disclosed prostate cancer-related molecules can include CEACAM5 (e.g., SEQ ID NOS: 21-26). In some embodiments, one of the disclosed prostate cancer-related molecules can include CEACAM6 (e.g., SEQ ID NOS: 27-31). In some embodiments, one of the disclosed prostate cancer-related molecules can include IL24 (e.g., SEQ ID NOS: 32-37). In some embodiments, one of the disclosed prostate cancer-related molecules can include MMP9 (e.g., SEQ ID NOS: 38-44). In some embodiments, one of the disclosed prostate cancer-related molecules can include CXCL14 (e.g., SEQ ID NOS: 45-50). In some embodiments, one of the disclosed prostate cancer-related molecules can include CD90 (e.g., SEQ ID NOS: 51-55). In some embodiments, one of the disclosed prostate cancer-related molecules can include POSTN (e.g., SEQ ID NOS: 56-61). In some embodiments, one of the disclosed prostate cancer-related molecules can include SFRP4 (e.g., SEQ ID NOS: 62-67). In some embodiments, one of the disclosed prostate cancer-related molecules can include PSA (e.g., SEQ ID NOS: 68-71). In some examples, combinations of these prostate cancer-related molecules are used, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13 of these.

Molecules that are similar to the prostate cancer-related molecules disclosed above can be used as well as fragments thereof that retain biological activity. These molecules may contain variations, substitutions, deletions, or additions (e.g., the variation carbamidomethyl cysteine may be used instead of cysteine). The differences can be in regions not significantly conserved among different species. Such regions can be identified by aligning the amino acid sequences of related proteins from various animal species. Generally, the biological effects of a molecule are retained. For example, a molecule at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to one of these molecules can be utilized. Molecules are of use that include at most 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 conservative amino acid substitutions. Generally, molecules are of use provided they retain at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% of the biological function of the native molecule, or have increased biological function as compared to the native molecule.

Administration of Therapy

Subjects analyzed with the disclosed methods and who are found to have prostate cancer can be selected for treatment. For example, subjects with prostate cancer or with intermediate-to-high-risk prostate cancer found to have increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 can be administered therapy for prostate cancer. Currently, the standard of care for prostate cancer can vary, but level of risk can be a factor. For example, a subject may be found to have low-risk prostate cancer, such as a patient with increased levels of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 but with MMP9 levels expected from a patient with low-risk prostate cancer. In some examples, subjects with low-risk prostate cancer may be treated using watchful waiting or active surveillance, both of which entail monitoring the cancer for changes and the subject for symptoms. Given that more invasive treatments entail a greater potential for side effects, studies suggest that active surveillance is the best choice for patients with low-risk prostate cancer.

In other examples, surgical removal of the prostate can be a treatment for low-risk prostate cancer or prostate cancers that do not respond to radiation therapy. In additional examples, subjects with any stage of prostate cancer, such as subjects with increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 can be treated with radiation therapy, such as using ionizing radiation to kill prostate cancer cells. In some other examples, subjects with either low- or intermediate-risk prostate cancer can be treated using brachytherapy, for example, where small radioactive particles, such as iodine-125 or palladium-103, are directly injected into the tumor, providing localized X-rays at the site of the tumor. In additional examples, ultrasound, such as high-intensity focused ultrasound (HIFU) is used where a subject has a recurrent case of prostate cancer, such as where a subject was successfully treated for prostate cancer but subsequently had increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4.

In further examples, a subject can be treated with hormone therapy, such as by modulating the levels of testosterone in the body, where the subject has either recurrent prostate cancer, for example, a subject that was successfully treated for prostate cancer but subsequently had increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4, or a subject that has high-risk prostate cancer, for example, a subject that has increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4 and increased expression of MMP9 compared with the expression expected from a patient with low-risk prostate cancer (e.g., as compared to a threshold of expression of any of these molecules established from one or more subjects, such as a cohort of control subjects).

In some examples, at least a portion of the prostate cancer is surgically removed (for example via cryotherapy), irradiated, chemically treated (for example via chemoembolization), or combinations thereof, as part of the therapy. For example, a subject having prostate cancer can have all or part of the tumor surgically excised prior to administration of additional therapy.

Exemplary agents that can be used include one or more anti-neoplastic agents, such as radiation therapy, chemotherapeutic, biologic (e.g., immunotherapy), and anti-angiogenic agents or therapies. Methods and therapeutic dosages of such agents are known to those skilled in the art, and can be determined by a skilled clinician. These therapeutic agents (which are administered in therapeutically effective amounts) and treatments can be used alone or in combination. In some examples, 1, 2, 3, 4 or 5 different anti-neoplastic agents are used as part of the therapy.

In one example the therapy includes administration of one or more chemotherapy immunosuppressants (such as Rituximab, steroids) or cytokines (such as GM-CSF). Chemotherapeutic agents are known (see for example, Slapak and Kufe, Principles of Cancer Therapy, Chapter 86 in Harrison's Principles of Internal Medicine, 14th edition; Perry et al., Chemotherapy, Ch. 17 in Abeloff, Clinical Oncology 2nd ed., 2000 Churchill Livingstone, Inc; Baltzer and Berkery. (eds): Oncology Pocket Guide to Chemotherapy, 2nd ed. St. Louis, Mosby-Year Book, 1995; Fischer Knobf, and Durivage (eds): The Cancer Chemotherapy Handbook, 4th ed. St. Louis, Mosby-Year Book, 1993). Exemplary chemotherapeutic agents that can be used with the therapy include but are not limited to, carboplatin, cisplatin, paclitaxel, docetaxel, doxorubicin, epirubicin, cabaziatxel, estramustine, vinblastine, topotecan, irinotecan, gemcitabine, iazofurine, etoposide, vinorelbine, tamoxifen, valspodar, cyclophosphamide, methotrexate, fluorouracil, mitoxantrone, and Doxil® (liposome encapsulated doxiorubicine). In one example the therapy includes docetaxel and prednisone. In one example the therapy includes cabaziatxel.

In one example, the therapy includes administering one or more of a microtubule binding agent, DNA intercalator or cross-linker, DNA synthesis inhibitor, DNA and/or RNA transcription inhibitor, antibodies, enzymes, enzyme inhibitors, and gene regulators.

Microtubule binding agents interact with tubulin to stabilize or destabilize microtubule formation thereby inhibiting cell division. Examples of microtubule binding agents that can be used as part of the therapy include, without limitation, paclitaxel, docetaxel, vinblastine, vindesine, vinorelbine (navelbine), the epothilones, colchicine, dolastatin 10, nocodazole, and rhizoxin. Analogs and derivatives of such compounds also can be used. For example, suitable epothilones and epothilone analogs are described in International Publication No. WO 2004/018478. Taxoids, such as paclitaxel and docetaxel, as well as the analogs of paclitaxel taught by U.S. Pat. Nos. 6,610,860; 5,530,020; and 5,912,264 can be used.

The following classes of compounds can be used as part of the therapy: suitable DNA and/or RNA transcription regulators, including, without limitation, anthracycline family members (for example, daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, and valrubicin) and actinomycin D, as well as derivatives and analogs thereof. DNA intercalators and cross-linking agents that can be administered to a subject include, without limitation, platinum compounds (for example, carboplatin, cisplatin, oxaliplatin, and BBR3464), mitomycins, such as mitomycin C, bleomycin, chlorambucil, cyclophosphamide, as well as busulfan, dacarbazine, estramustine, and temozolomide and derivatives and analogs thereof. DNA synthesis inhibitors suitable for use as therapeutic agents include, without limitation, methotrexate, 5-fluoro-5′-deoxyuridine, 5-fluorouracil and analogs thereof. Examples of suitable enzyme inhibitors include, without limitation, camptothecin, etoposide, exemestane, trichostatin and derivatives and analogs thereof. Suitable compounds that affect gene regulation include agents that result in increased or decreased expression of one or more genes, such as raloxifene, 5-azacytidine, 5-aza-2′-deoxycytidine, tamoxifen, 4-hydroxytamoxifen, mifepristone, and derivatives and analogs thereof. Kinase inhibitors include imatinib, gefitinib, and erolitinib that prevent phosphorylation and activation of growth factors.

In one example, the therapy includes folic acid (for example, methotrexate and pemetrexed), purine (for example, cladribine, clofarabine, and fludarabine), pyrimidine (for example, capecitabine), cytarabine, fluorouracil, gemcitabine, and derivatives and analogs thereof. In one example, the therapy includes a plant alkaloid, such as podophyllum (for example, etoposide) and derivatives and analogs thereof. In one example, the therapy includes an antimetabolite, such as cytotoxic/antitumor antibiotics, bleomycin, hydroxyurea, mitomycin, and derivatives and analogs thereof. In one example, the therapy includes a topoisomerase inhibitor, such as a topoisomerase I inhibitor (e.g., topotecan, irinotecan, indotecan, indimitecan, camptothecin and lamellarin D) or a topoisomerase II inhibitor (e.g., etoposide, doxorubicin, daunorubicin, mitoxantrone, amsacrine, ellipticines, aurintricarboxylic acid, ICRF-193, genistein, and HU-331), and derivatives and analogs thereof. In one example, the therapy includes a photosensitizer, such as aminolevulinic acid, methyl aminolevulinate, porfimer sodium, verteporfin, and derivatives and analogs thereof. In one example, the therapy includes a nitrogen mustard (for example, chlorambucil, estramustine, cyclophosphamide, ifosfamide, and melphalan) or nitrosourea (for example, carmustine, lomustine, and streptozocin), and derivatives and analogs thereof.

Other therapeutic agents, for example anti-tumor agents, that may or may not fall under one or more of the classifications above, also are suitable for therapy. By way of example, such agents include adriamycin, apigenin, rapamycin, zebularine, cimetidine, amsacrine, anagrelide, arsenic trioxide, axitinib, bexarotene, bevacizumab, bortezomib, celecoxib, estramustine, hydroxycarbamide, lapatinib, pazopanib, masoprocol, mitotane, tamoxifen, sorafenib, sunitinib, vandetanib, tretinoin, and derivatives and analogs thereof.

In one example, the therapy includes one or more biologics, such as a therapeutic antibody, such as monoclonal antibodies. Examples of such biologics that can be used include one or more of bevacizumab, cetuximab, panitumumab, pertuzumab, trastuzumab, bevacizumab (Avastin®), ramucirumab, and the like. In specific examples, the antibody or small molecules used as part of the therapy include one or more of the monoclonal antibodies cetuximab, panitumumab, pertuzumab, trastuzumab, bevacizumab (Avastin®), ramucirumab, or a small molecule inhibitor such as gefitinib, erlotinib, and lapatinib.

In some examples the therapy includes administration of one or more immunotherapies, which may include the biologics listed herein. In specific examples, the immunotherapy includes therapeutic cancer vaccines, such as those that target PSA (e.g., ADXS31-142), prostatic acid phosphatase (PAP) antigen, TARP, telomerase (e.g., GX301) or that deliver 5T4 (e.g., ChAdOx1 and MVA); antigens NY-ESO-1 and MUC1; antigens hTERT and survivin; prostate-specific antigen (PSA) and costimulatory molecules (e.g., LFA-3, ICAM-1, and B7.1) directly to cancer cells, such as rilimogene galvacirepvac. Other examples of therapeutic vaccines include DCVAC, sipuleucel-T, pTVG-HP DNA vaccine, pTVG-HP, JNJ-64041809, PF-06755992, PF-06755990, and pTVG-AR. In other examples, the immunotherapy includes oncolytic virus therapy, such as aglatimagene besadenovec, HSV-tk, and valacyclovir. In additional examples, the immunotherapy can include checkpoint inhibitors, such as those that target PD-1 (e.g., nivolumab, pembrolizumab, durvalumab, and atezolizumab), CTLA-4 (e.g., tremelimumab and ipilimumab), B7-H3 (e.g., MGA271), and CD27 (e.g., CDX-1127). The protein MGD009 may also be used in another example. In specific examples, the immunotherapy can also include adoptive cell therapy, such as those that include T cells engineered to target NY-ESO-1 and those that include natural killer (NK) cells. In some examples, the immunotherapy can include adjuvant immunotherapies, such as sipuleucel-T, indoximod, and mobilan. In other specific examples, the immunotherapy includes one or more of tisotumab vedotin, sacituzumab govitecan, LY3022855, BI 836845, vandortuzumab vedotin, and BAY2010112, and MOR209/ES414. In additional examples, the immunotherapy can include cytokines, such as CYT107, AM0010, and IL-12.

In some examples, the subject receiving the therapy is also administered interleukin-2 (IL-2), as part of the therapy, for example via intravenous administration. In particular examples, IL-2 is administered at a dose of at least 500,000 IU/kg as an intravenous bolus over a 15 minute period every eight hours beginning on the day after administration of the peptides and continuing for up to 5 days. Doses can be skipped depending on subject tolerance.

In some examples, the subject receiving the therapy is also administered a fully human antibody to cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4) as part of the therapy, for example via intravenous administration. In some example subjects receive at least 1 mg/kg anti-CTLA-4 (such as 3 mg/kg every 3 weeks or 3 mg/kg as the initial dose with subsequent doses reduced to 1 mg/kg every 3 weeks).

In one specific example for a subject with prostate cancer, such as a subject with increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4, the therapy can include one or more of abiraterone acetate, bicalutamide, cabazitaxel, casodex (bicalutamide), degarelix, docetaxel, enzalutamide, flutamide, goserelin acetate, jevtana (cabazitaxel), leuprolide acetate, lupron (leuprolide acetate), lupron depot (leuprolide acetate), lupron depot-3 month (leuprolide acetate), lupron depot-4 month (leuprolide acetate), lupron depot-ped (leuprolide acetate), mitoxantrone hydrochloride, nilandron (nilutamide), nilutamide, provenge (sipuleucel-t), radium 223 dichloride, sipuleucel-T, taxotere (docetaxel), viadur (leuprolide acetate), xofigo (radium 223 dichloride), xtandi (enzalutamide), zoladex (goserelin acetate), and zytiga (abiraterone acetate).

In another specific example for a subject with prostate cancer, such as a subject with increased expression of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4, the therapy can include one or more of chemotherapy drugs, such as cabazataxel (Jevtana®), docetaxel (Taxotere®), mitoxantrone (Teva®), or androgen deprivation therapy (ADT), such as with abiraterone Acetate (Zytiga®), bicalutamide (Casodex®), buserelin Acetate (Suprefact®), cyproterone Acetate (Androcur®), degarelix Acetate (Firmagon®), enzalutamide (Xtandi®), flutamide (Euflex®), goserelin Acetate (Zoladex®), histrelin Acetate (Vantas®), leuprolide Acetate (Lupron®, Eligard®), triptorelin Pamoate (Trelstar®). The therapy can also include drugs to treat bone metastases (bisphosphate therapy), such as alendronate (Fosamax®), denosumab (Xgeva®), pamidronate (Aredia®), zoledronic acid (Zometa®), or radiopharmaceuticals, such as radium 223 (Xofigo®), strontium-89 (Metastron®), and samarium-153 (Quadramet®).

The therapy can be administered in cycles (such as 1 to 6 cycles), with a period of treatment (usually 1 to 3 days) followed by a rest period. But some therapies can be administered every day.

EXAMPLE 1 Methods and Materials

This example provides technical details and procedures, including relevant instrument settings and materials, used to obtain the protein expression data from patient urine samples discussed in the Examples below.

Urine Collection, Processing, and Protein Digestion

Use of human urine samples was approved by an Institutional Review Boards with written consent of donors, and the samples were anonymized before being given to researchers. The suffix N was added to the sample codes to denote non-cancer, and the suffix C to denote cancer from pre-operative (pre-op) patients. Urine from post-operative patients (post-op) was collected after surgical resection of the prostate. All urine samples were freshly collected for this study; no archived samples were used.

Collected voided urine samples were processed within 2 h (to isolate RNA as well). The samples were centrifuged at 1,200 rpm for 5 min, and the supernatant was stored at −80° C. Fifteen to 100 mL of urine was desalted and concentrated using Amicon® Ultra-15 (3 kDa molecular weight cut-off, Millipore, USA). Protein concentrations were determined by the BCA assay (Pierce, USA). Concentrated urinary proteins from each sample, ranging from 200 to 300 μg, were denatured and reduced with 8 M urea and 10 mM DTT in 50 mM NH4HCO3, pH 8.0 for 1 h at 37° C. Protein cysteine residues were alkylated with 40 mM iodoacetamide for 1 h at room temperature in the dark. The resulting sample was diluted 6-fold with 50 mM NH4HCO3, pH 8.0, and digested by sequencing-grade modified porcine trypsin (Promega, USA) at 1:50 trypsin:protein (w/w) overnight at 37° C. The resulting digest was desalted by using a 1 mL SPE C18 column (Supelco, USA) as described previously (Shi et al., J Proteome Res 13:875-82, 2014). The final tryptic peptide concentration was determined by the BCA assay. The peptide sample was diluted to 0.5 μg/μL with 0.1% formic acid in water, and heavy isotope-labeled synthetic peptides were spiked in at an equimolar concentration, 10 fmol/μt of crude heavy peptides for the secreted protein markers, 10 fmol/μt of pure heavy peptide IVGGWECcamEK (Ccam: cysteine residue synthesized as carbamidomethyl cysteine), and 1 fmol/μt of pure heavy peptide LSEPAELTDAVK for PSA, due to the response difference between the two PSA peptides.

Database Query

The human urine proteome databases archived in PeptideAtlas (www.peptideatlas.org) were queried for data entries of marker identifiers. The UrinePA build contained high confidence peptide and protein identifications obtained from five labs using tandem mass spectrometry proteomics (Farrah et al., J Proteome Res 13:60-75, 2014). About 2,500 non-redundant proteins were cataloged at a 1% false discovery rate. Another database listed 587 entries of a “Core Urinary Proteome”, which was established from an in-depth analysis of second morning urine obtained over three days from seven healthy volunteers between 25-35 years old (Nagaraj and Mann, J Proteome Res 10:637-45, 2011).

Chemical Reagents

Urea, dithiothreitol (DTT), iodoacetamide, ammonium formate, trifluoroacetic acid (TFA), and formic acid were purchased from Sigma (USA). The synthetic peptides labeled with 13C/15N on C-terminal lysine and arginine residues were from Thermo Scientific (USA). The heavy peptides for PSA protein were estimated to be of >95% purity by HPLC.

SRM Assays

Ten surrogate peptides were first chosen for the protein markers based on in silico trypsin digestion and existing MS/MS data, the Global Proteome Machine (GPM), and PeptideAtlas. These peptides were then evaluated by ESP predictor (Fusaro et al., Nat Biotechnol 27:190-8, 2009) and CONSeQuence software (Eyerset al., Mol Cell Proteomics 10:M110.003384, 2011). Three to five peptides with moderate hydrophobicity and high scores from the prediction tools were selected for peptide synthesis. The synthesized crude heavy-isotope labeled peptides were further evaluated for peptide response and fragmentation pattern. Optimal collision energy (CE) values were achieved by direct infusion of the individual peptides and/or multiple LC-SRM runs with CE ramping. The best performing peptides were used for detection and quantification of the secreted protein markers. For each peptide, the three best transitions and matrix interference were determined. The relative intensity ratios among the three selected transitions for SRM were predefined by the internal standard heavy peptides in buffer. The matrix interference for a given transition that fell into mass widths Q1 and Q3 from co-eluting peptides was identified by a deviation from the expected relative intensity ratios among the transitions. The transition with no matrix interference was used for marker quantification in urine samples.

LG-SRM Measurement

The LG-SRM approach was previously demonstrated to enable reproducible quantification of target proteins at ˜10 ng/mL levels in nondepleted human serum (Shi et al., Anal Chem 85:9196-203, 2013). Typically, 4 μL of the tryptic digest samples with a peptide concentration of 0.5 μg/μL was directly loaded onto a capillary reversed-phase column, 75 μm inner diameter (i.d.)×150 cm length, packed in-house with 3-μm Jupiter C18 bonded particles (Phenomenex, USA) to permit long gradient separation without a trap column with its dead volume affecting peptide retention time. Peptide separations were performed at a mobile phase flow rate of 100 nL/min on a binary pump system using 0.1% formic acid in water as phase A and 0.1% formic acid in 90% acetonitrile as phase B. The profile for a 300-min gradient time was 5-15% B in 27 min, 15-25% B in 140 min, 25-35% B in 73 min, and 35-90% B in 60 min. The TSQ Vantage mass spectrometer was operated in the manner as previously described (Shi et al., Anal Chem 85:9196-203, 2013).

PRISM-SRM Measurement

The PRISM-SRM approach has been described for quantification of low-abundance proteins in human plasma or serum (Shi et al., Proc Natl Acad Sci USA 109:15395-400, 2012). Briefly, high-resolution reversed-phase capillary LC with pH 10 mobile phase was used as the first dimensional separation of peptides from trypsin-digested human urine proteins. Following separation, the column eluent was automatically collected every minute into a 96-well plate during a ˜100-min LC run while on-line SRM monitoring of heavy internal standard peptides was performed on a small split stream of the flow. Intelligent selection (termed iSelection) of target peptide fractions was achieved based on the on-line SRM signal of internal standard peptides. Prior to peptide fraction collection, 17 μL of water was added to each well to minimize excessive loss of peptides and dilute the peptide fractions (˜1:7) for LC-SRM analysis.

Following iSelection, the target peptide-containing fractions were subjected to LC-SRM measurement. All peptide fractions were analyzed by using the nanoACQUITY UPLC® system (Waters Corporation, USA) coupled on-line to a TSQ Vantage triple quadrupole mass spectrometer (Thermo Scientific). Solvents used were 0.1% formic acid in water (mobile phase A) and 0.1% formic acid in 90% acetonitrile (mobile phase B). Peptide separations were performed at a mobile phase flow rate of 400 nL/min using an ACQUITY UPLC BEH 1.7 μm C18 column (75 μm i.d.×10 cm), which was connected to a chemically etched 20 μm i.d. fused-silica emitter via a Valco stainless steel union. Four microliters of individual peptide fractions (total volume 20 μL) following PRISM were injected for LC separation using a binary gradient of 10-20% phase B in 7 min, 20-25% phase B in 17 min, 25-40% phase B in 1.5 min, 40-95% phase B in 2.5 min, and 95% phase B in 6 min for a total time of ˜35 min. The TSQ Vantage was operated in the same manner as described (Shi et al., J Proteome Res 13:875-82, 2014). A scan width of 0.002 m/z and a dwell time of 40 ms were set for all SRM transitions.

Proteomics Data Analysis

SRM data were analyzed using Skyline software (MacLean et al., Bioinformatics 26:966-8, 2010). Peak detection and integration were determined based on two criteria: (1) the same retention time and (2) approximately the same relative SRM peak intensity ratios across multiple transitions between light (L) peptides and heavy (H) peptide standards (Shi et al., J Proteome Res 13:875-82, 2014; Shi et al., J Proteome Res12:3353-61, 2013]. All data were manually inspected to ensure correct peak detection and accurate integration. The signal to noise ratio (S/N) was calculated by the peak apex intensity over the highest background noise in a retention time region of ±15 s for the target peptides (Shi et al., Proc Natl Acad Sci USA 109:15395-400, 2012; Shi et al., J Proteome Res 12:3353-61, 2013). The background noise levels were conservatively estimated by visually inspecting chromatographic peak regions. Quantifiable endogenous surrogate peptides should have SRM signals with S/N≥10. The RAW data from the TSQ Vantage were loaded into Skyline to create high resolution figures of extracted ion chromatograms (XICs) of multiple transitions monitored for the target peptides/proteins.

Statistical Analysis

GraphPad Prism 6.0 was used for statistical analysis and plotting; P<0.05 was considered statistically significant. Receiver operating characteristic (ROC) curves were produced in terms of sensitivity and specificity of protein markers at their specific cutoff values. The optimal cutoff was the point with the best sum of sensitivity and specificity. Multivariate evaluative analysis for various combinations of protein markers was performed using SPSS 16 by logistic regression to find the best-fitting model for each comparison group.

AGR2 ELISA

Urinary AGR2 was measured by a developed sandwich ELISA based on monoclonal antibodies P1G4 (IgG1) to capture and P3A5 (IgG2a) to detect as described (Wayner et al., Prostate 72:1023-34, 2012). Recombinant AGR2 (GenWay Biotech, San Diego, Calif.) was used to generate a standard curve. Purified P1G4 at 1:1,000 was used to capture the analyte, and purified P3A5 at 1:1,000 was used for detection followed by HRP-conjugated anti-mouse IgG2a (SouthernBiotech, USA). The chromogenic reaction was recorded at plate reader setting of λ=405 nm. Culture media of the AGR2-secreting prostate cancer cell line PC3 was used as a positive control. Buffer was the negative control.

Calculations

  • Secreted protein concentration calculation in urine (ng/100 μg of total urinary protein):


[secreted protein]=L/H×IS (fmol/μL)×MW×10−6(ng/fmol)×200 μL

  • IS: internal standard
  • MW: protein molecular weight (g/mol)
  • 100 μg of urinary protein: 200 μL of 0.5 μg/μL urine peptide

EXAMPLE 2 Tumor-Associated Secreted Proteins in Human Urine

This example describes methods that were used to determine the tumor-associated secreted proteins present in urine.

Through comparison of cell type-specific transcriptomes, genes showing elevated tumor expression and encoding secreted/extracellular proteins were identified from both the epithelial and stromal compartments. Furthermore, gene expression analysis indicated that many showed differential expression among tumors of different Gleason scores. The epithelial-derived marker candidates included AGR2, AGR3, CRISP3, CEACAM5, CEACAM6, CCL3, CCL4, IL24, and MMP9; the stromal-derived candidates included CXCL14, CD90, IL24, MMP9, POSTN, SFRP4, and WISP1. The database PeptideAtlas was interrogated to determine if these proteins were present in non-cancer urine. The query result is shown in FIG. 1. In the UrinePA archive of available mass spectrometry datasets, the “observed” qualifier was used to indicate protein abundance. UMOD and ALB were the most abundant with the observed times 24,115 and 33,149, respectively. Other proteins, such as APOD (2,352), LMAN2 (639), AMBP (15,223), F2 (1,164), PTGDS (4,479), and AZGP1 (1,903), were the major urinary proteins that could be visualized by staining of a polyacrylamide gel electrophoregram. Prostatic proteins, such as KLK3/PSA (193), KLK2 (2), ACPP (415), MSMB (21), PSCA (123), and PRSS8 (230), were also cataloged as was stromal cell PENK (6). ERG (0), a cytoplasmic protein in prostate cancer cells and endothelial cells, was not found. Non-secreted UPK3A (3) was present for bladder cells. A group of cell surface CD antigens expressed by luminal and cancer cells (CD26, CD10, CD57, CD38, CD107a, and CD107b) were also found. CD45 (0) was queried for leukocytes, and CD31 (9) for endothelial cells. Of the marker candidates, CRISP3 (65), CEACAM5 (21), CEACAM6 (5), THY1/CD90 (261), MMP9 (115), and SFRP4 (17) were cataloged in non-cancer, demonstrating that they could be detected in all urine samples. Therefore, those proteins that were not encountered in the database of healthy donors were more likely derived from diseased tissues, such as prostate cancer in the urinary system.

EXAMPLE 3 Multiplexed Peptide Assays for Prostate Cancer Protein Markers

This example describes methods that were used to develop sensitive multiplexed peptide assays for prostate cancer-associated protein markers.

Selection of the most suitable surrogate peptides is important for sensitive accurate quantification of target proteins in patient specimens. For each prostate cancer protein marker, the surrogate peptides selected for assay development are listed in FIG. 2. For PSA, IVGGWECcamEK (SEQ ID NO: 70) and LSEPAELTDAVK (SEQ ID NO: 71) were demonstrated to be most effective for quantification (Shi et al., Proc Natl Acad Sci USA 109:15395-400, 2012; Keshishian et al., Mol Cell Proteomics 6:2212-29, 2007). For the others, a pooled prostate cancer patient urine sample was used to configure the final SRM assays with an evaluation of matrix interference, endogenous peptide detectability, and the peptide SRM response. LG-SRM was used first to simultaneously measure the 14 candidates due to its moderate sensitivity (≥10-fold higher than LC-SRM) and higher multiplexing capability (˜three times higher than LC-SRM) (Shi et al. Anal Chem 85:9196-203, 2013). PSA, CD90 (THY1), CRISP3, CXCL14, IL24, MMP9, POSTN, and SFRP4 (note protein names of these genes, which could be different, are given in the figures), were confidently detected and quantified by at least one surrogate peptide (FIGS. 3A-3B and 4). The ultrasensitive PRISM-SRM (≥20-fold higher in sensitivity than LG-SRM) was then used to measure the other seven candidates. Five proteins, AGR2, AGR3, CCL3, CEACAM5, and CEACAM6, were reliably detected and quantified (FIGS. 3A-3B). The reproducibility of LG-SRM and PRISM-SRM based assays for measurements in biofluids such as urine and serum has been validated and typically exhibit a coefficient of variance (CV) <10%. Based on the LG-SRM and PRISM-SRM results, SRM assays were established for each of the detectable peptides with the three best transitions without matrix interference, and the best transition was used for quantification (FIG. 4). From the assay results, the 12 detected markers were grouped into seven moderate-to-low abundance proteins for LG-SRM analysis and five low abundance proteins for PRISM-SRM analysis. The two undetected proteins, CCL4 and WISP1, were excluded from further testing. The configured SRM assays were used for measuring the 12 protein biomarkers in a cohort of 14 cancer (pre-op) and 6 non-cancer (healthy control) urine samples. Among the 12 proteins, after excluding CCL4 and WISP1, the remaining 10 proteins can be reliably detected and quantified across the 20 urine subjects with at least one surrogate peptide without co-eluting interference. Another cohort of post-op urine samples was collected and analyzed separately to determine the origin of urinary PSA.

EXAMPLE 4 Quantification Accuracy of Individual Surrogate Peptides

This example shows assessment of the quantification accuracy for individual surrogate peptides to determine which surrogate peptides are suitable for diagnosis.

Multiple surrogate peptides were tested for quantification of a specific protein in urine. Without posttranslational modifications or undocumented amino acid changes in the surrogate peptides, their measured concentrations should have a high degree of correlation across all samples because the surrogate peptide level was stoichiometric to that of their cognate protein (Worboys et al., Nat Methods 11:1041-4, 2014). With any changes in the peptide sequence, the level of the unmodified surrogate peptides would be lower, affecting accurate measurement of their corresponding proteins. For these differences, each surrogate peptide could potentially represent a distinctive signature with diagnostic value (Zhang et al., J Exp Med 210:191-203, 2013).

To evaluate the quantification accuracy of surrogate peptides, a correlation analysis of the L/H ratios between the surrogate peptides from the same protein was carried out. MMP9 was represented by four quantifiable surrogate peptides, and the Pearson correlation coefficients ranged from 0.59 between FQTFEGDLK (SEQ ID NO: 41) and SLGPALLLLQK (SEQ ID NO: 43) to 0.93 between AVIDDAFAR (SEQ ID NO: 40) and FQTFEGDLK (SEQ ID NO: 41), which indicated that multiple MMP9 isoforms could exist in these clinical urine samples (FIGS. 5A-5F). For CD90, low correlation coefficients between VTSLTACLVDQSLR (SEQ ID NO: 54) and the other two peptides were obtained, whereas a good correlation, R2=0.72, was obtained with the other two peptides (FIGS. 6A-6B). These data show that unknown modifications were present in the VTSLTACLVDQSLR (SEQ ID NO: 54) sequence in several urine samples, making this peptide unsuitable for quantitative measurement of CD90. However, such modifications could be cancer-specific.

A high correlation between the two widely used PSA peptides, IVGGWECcamEK (SEQ ID NO: 70) and LSEPAELTDAVK (SEQ ID NO: 71), was obtained with R2=0.99 across the 20 urine samples (FIGS. 7A-7B). Closer examination revealed three data points from urine samples P08-015C, P07-040C, and P06-017C that deviated from the correlation plot (FIGS. 7A-7B and FIG. 8). These data show that the two PSA peptides in these urine samples contained amino acid alterations. For example, the P06-017C urine showed a lower L/H ratio for LSEPAELTDAVK (SEQ ID NO: 71). The experimental results indicate an amino acid change in the LSEPAELTDAVK sequence (FIG. 9; SEQ ID NO: 71). These data are supported by a recent discovery of PSA proteoforms, a nonsynonymous mutation L1321 (rs2003783) within LSEPAE(L/I)TDAVK (SEQ ID NO: 71) (Végvári et al., Mol Cell Proteomics 12:2761-73, 2013). For the other two urine samples with the lower L/H ratios for IVGGWECcamEK (SEQ ID NO: 70), a similar type of change or others could be present (Mikolajczyk et al., Urology 59:797-802, 2002).

EXAMPLE 5 Urinary PSA Exclusively Secreted by the Prostate Gland

This example shows that urinary PSA is primarily secreted by the prostate gland and that its secretion from other sources is negligible.

To quantitatively evaluate the percentage of urinary PSA originating from the prostate gland, LC-SRM was used to measure its concentration in seven post-op urine samples (i.e., from men without a prostate) and the cohort of 20 urine samples before radical prostatectomy (FIG. 8). The measured PSA levels ranged from 0.02 ng/100 μg to 2.95 ng/100 μg of total urinary protein with an average value of 0.98 ng/100 μg (and a median value of 0.41 ng/100 μg, FIG. 10). When compared to the PSA levels in the non-cancer and cancer samples with an average value of 110.89 ng/100 μg of total urinary protein (and a median value of 28.68 ng/100 μg), the percentage of PSA in the post-op urine was only ˜1% (˜1.5% median value, FIG. 10). In non-cancer and cancer, urinary PSA can be contributed by all possible sources, whereas in post-op, it cannot be contributed from an absent prostate gland. Thus, the data show that urinary PSA is secreted mostly from the prostate gland, and contribution from other sources in the urinary system is negligible.

EXAMPLE 6 Normalizing the Concentration of Cancer-Associated Proteins Secreted Using PSA

This example shows normalization of cancer-associated secreted protein concentration using PSA to facilitate differentiation of cancer and non-cancer samples.

In the SRM targeted measurement, the reported L/H peak area ratios of surrogate peptides were proportional to the concentrations of their cognate protein and are expressed as ng/100 μg of total urinary protein because the same amount of the heavy internal standards was added to the digested peptide mixtures for all 20 samples, which have the same peptide concentrations. Thus, the L/H ratio is the adjusted concentration of the target protein in urine (against the total amount of the urinary proteins, Shi et al., J Proteome Res 13:875-82, 2014, FIGS. 11A-11B). This adjustment accounts for substantial variations in urinary protein concentration among donors and sometimes among donations from the same donor. For most surrogate peptides measured, the cancer urine showed higher median L/H values than the non-cancer urine; while for several other peptides (CRISP3, CXCL14, IL24, and SFRP4), a lower or equal median L/H value in cancer vs. non-cancer was found. A Mann-Whitney U test of the surrogate peptide L/H ratios revealed no significant difference between cancer and non-cancer for all of the secreted protein markers (FIG. 12). Because the secreted urinary proteome is a pool comprised of all proteins produced by organs along the urinary tract (see PeptideAtlas query result), “normalization” to the secreted proteins solely produced by the prostate was necessary. The post-op urine analysis showed that PSA levels, which were proportional to the size of the gland and the number of secretory cells, luminal and cancer, can be used for this normalization. PSA normalization was used to present urinary AGR2 levels as AGR2/PSA ratios (Shi et al., J Proteome Res 13:875-82, 2014). The amount of PSA was similarly used in the urine PCA3 assay in which the urinary PCA3 score was generated by normalizing the PCA3 transcript levels to those of PSA transcript (Marks and Bostwick, Rev Urol 10:175-81, 2008).

Based on the above peptide correlation analysis, both PSA peptides could be used to determine PSA protein concentrations in the cohort. The secreted protein marker/PSA concentration ratios were obtained by dividing the L/H peak area ratios of surrogate peptides for the protein markers by the L/H peak area ratio of the PSA peptide IVGGWECcamEK (SEQ ID NO: 70) (FIGS. 13A-13B). After PSA normalization, a significant difference between the cancer and non-cancer urine was observed for the marker peptides [except for LPQTLSR (SEQ ID NO: 3) of AGR2 (see below), VTSLTACLVDQSLR (SEQ ID NO: 54) of CD90, and MVIITTK (SEQ ID NO: 47) of CXCL14] with the P values ranging from 0.015 to 0.035 (FIG. 12 and FIGS. 14A-14F). These results demonstrate the utility of PSA protein normalization in prostate cancer urine biomarker performance.

ROC analyses with 95% confidence intervals show that the peptides with P<0.05 produced AUC values>0.80, while for the three peptides with P>0.05, the AUC values produced were <0.80 (FIG. 12 and FIGS. 14A-14B). With PSA normalization, all the peptide signatures showed values of the sum of sensitivity and specificity ranging from 1.60 to 1.79 at the optimal cutoff points. These data show good biomarker performance in differentiation of prostate cancer from non-cancer through urine analysis. In addition, surrogate peptides with a good correlation produced the same AUC values. For example, the AUC values obtained from the four MMP9 surrogate peptides, AVIDDAFAR (SEQ ID NO: 40), FQTFEGDLK (SEQ ID NO: 41), LGLGADVAQVTGALR (SEQ ID NO: 42), and SLGPALLLLQK (SEQ ID NO: 43), were 0.82, 0.86, 0.86, and 0.86, respectively. The AUC values from the two well-correlated CD90 surrogate peptides, VLYLSAFTSK (SEQ ID NO: 53) and HVLFGTVGVPEHTYR (SEQ ID NO: 55), were 0.86 and 0.87, respectively. The VTSLTACLVDQSLR peptide (SEQ ID NO: 54) without significant correlations produced an AUC value of 0.77 (FIG. 12). Multi-marker performance was also assessed by using a multivariate analysis of various combinations of the surrogate peptides (FIG. 15). The best combination consisted of LPQTLSR (SEQ ID NO: 3) from AGR2, LYTYEPR (SEQ ID NO: 6) from AGR3, SDLVNEEATGQFR (SEQ ID NO: 23) from CEACAM5, VTSLTACLVDQSLR (SEQ ID NO: 54) from CD90, and GVCISPEAIVTDLPEDVK (SEQ ID NO: 64) from SFRP4. With a P value of 0.002 and an AUC value of 0.95, this peptide grouping outperformed any single marker (FIGS. 14E-14F, FIG. 12, and FIG. 15).

EXAMPLE 7 Transient Increase in Urinary AGR2 in Non-Cancer Urine

This example demonstrates transient increases in urinary AGR2 in non-cancer urine and a persistently high urinary AGR2 in cancer urine.

A fraction of non-cancer urine was found to have an above-background amount of urinary AGR2, although urine proteome database query yielded no AGR2 identifier (cf. FIG. 1). The AGR2 levels in a middle-age non-cancer male (with normal serum PSA (sPSA)) over a two-week period are shown in FIG. 16B. The levels increased on day 4 (for both morning and afternoon urine) followed by a decrease to the baseline level on subsequent days. Treatment of urine samples by the addition of alcohol (to lyse shed AGR2+ bladder cells) did not raise the level of detectable urinary AGR2 (bars 3 vs. 4, FIG. 16B). Thus, some day-to-day physiological differences could account for AGR2 in the urine of non-cancer samples. This phenomenon affected the AUC for AGR2. Unlike non-cancer samples, AGR2 in cancer urine samples remained above the background, and the urine contained other marker proteins. Testing this hypothesis required a suitable patient donor to volunteer donations every day over a similar period of two weeks. Nevertheless, the AUC value obtained for AGR2 was 0.74 (FIG. 16A) by ELISA and 0.77 by PRISM-SRM. Hence, both biomarker measurement methods produced concordant results.

EXAMPLE 8 Detection of Clinically Significant Cancers by Secreted Protein Markers

This example compares differentiative power in identifying the low-risk from the high-risk cancers among secreted protein markers. This example also shows that MMP9 expression can be used to differentiate low- and high-risk prostate cancer.

The prostate cancer cohort was grouped into either low volume/low grade: Gleason score≤6 and dominant tumor volume≤0.5 cc (Nakanishi et al., J Urol 179:1804-9, 2008) or clinically significant: not meeting the criteria for the low volume/low grade disease. The marker levels for low-risk cancer and high-risk cancer were analyzed by Mann-Whitney's U test (FIGS. 17 and 18A-18B). For most surrogate peptides, the marker/PSA ratios were lower in men with low volume/low grade than those with high volume/high grade; thus, more cancer-associated proteins were produced and secreted into urine from significant cancers. However, most surrogate peptides did not show a significant differentiative power in identifying the low-risk from the high-risk cancers (FIGS. 18A-18B). One exception was MMP9. Two surrogate peptides, FQTFEGDLK (SEQ ID NO: 41) and LGLGADVAQVTGALR (SEQ ID NO: 42), produced a P value of 0.022 in comparing low volume/low grade cancer and significant cancer (FIGS. 18A-18B and FIG. 19A). The AUC values obtained from an ROC analysis for the two MMP9 peptides were above 0.90; thus, MMP9 and Gleason and tumor volume were associated in this patient cohort. This result is supported by the dataset query of cancer cell type transcriptomes. The array signal intensity value for MMP9 in Gleason pattern 4 cancer cells is 3004.10, 12-fold higher than 238.41 in Gleason pattern 3 cancer cells. (FIGS. 20A-20B). Other secreted proteins, such as AGR2, showed lower array signal intensity values in Gleason 4 cancer cells than Gleason 3 cancer cells. For comparison, urine PSA and serum PSA showed no power in differentiating the two cancer types (FIGS. 19B-19C). This analysis was blinded because the pathology parameters were not made known before the quantitative SRM measurements.

In view of the many possible embodiments to which the principles of the disclosure may be applied, it should be recognized that illustrated embodiments are only examples of the disclosure and should not be considered a limitation on the scope of the invention. Rather, the scope of the invention is defined by the following claims. We therefore claim as our invention all that comes within the scope and spirit of these claims.

Claims

1. A method of treating a subject with prostate cancer, comprising:

measuring expression of at least two prostate cancer-related molecules in a sample obtained from a subject, wherein the prostate cancer-related molecules comprise at least two of anterior gradient 2 (AGR2), anterior gradient 3 (AGR3), cysteine-rich secretory protein 3 (CRISP3), chemokine (C-C motif) ligand 3 (CCL3), carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6), interleukin 24 (IL24), matrix metalloproteinase 9 (MMP9), CXC motif ligand 14 (CXCL14), cluster designation 90 (CD90), periostin (POSTN), and secreted frizzled-related protein 4 (SFRP4);
measuring increased expression of the at least two prostate cancer-related molecules as compared to a control representing expression for each of the at least two prostate cancer-related molecules expected in a sample from a subject who does not have prostate cancer; and
administering at least one of watchful waiting, active surveillance, surgery, radiation, hormone therapy, chemotherapy, brachytherapy, cryotherapy, ultrasound, bisphosphate therapy, biologic therapy, or vaccine therapy to the subject with prostate cancer, thereby treating the subject.

2. The method of claim 1, wherein the at least two prostate cancer-related molecules comprise:

all of AGR2, AGR3, CEACAM5, CD90, and SFRP4; or
all of AGR2, AGR3, CRISP3, CCL3, CEACAM5, CEACAM6, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4.

3. The method of claim 1, wherein the at least two prostate cancer-related molecules comprise a combination listed in FIG. 15.

4. The method of claim 1, wherein the at least two prostate cancer-related molecules are low-abundance molecules.

5. The method of claim 4, wherein the low-abundance molecules comprise AGR2, AGR3, CCL3, CEACAM5, and CEACAM6.

6. The method of claim 1, wherein the at least two prostate cancer-related molecules are moderate-to-low-abundance molecules.

7. The method of claim 6, wherein the moderate-to-low-abundance molecules comprise CRISPS, IL24, MMP9, CXCL14, CD90, POSTN, and SFRP4.

8. The method of claim 1, wherein the expression is determined based on protein concentration.

9. The method of claim 8, wherein the protein concentration is determined using the concentration of at least one surrogate peptide of the protein.

10. The method of claim 9, wherein the at least one surrogate peptide is a peptide listed in FIG. 4.

11. The method of claim 1, further comprising normalizing expression of the at least two prostate cancer-related molecules to a prostate protein concentration.

12. The method of claim 11, wherein normalizing to prostate protein concentration comprises normalizing to prostate specific antigen (PSA) concentration.

13. The method of claim 12, wherein normalizing to PSA concentration comprises normalizing to the concentration of at least one surrogate peptide of PSA.

14. The method of claim 13, wherein at least one surrogate peptide of PSA is IVGGWECEK (SEQ ID NO: 70) or LSEPAELTDAVK (SEQ ID NO: 71).

15. The method of claim 1, wherein the sample obtained from the subject is a urine sample.

16. The method of claim 1, wherein the subject is human.

17. The method of claim 1, wherein the detected expression of the at least two prostate cancer-related molecules has an AUC value of greater than 0.75.

18. A method, comprising:

treating a sample obtained from a subject with a protease, thereby forming a digested sample; and
measuring expression of AGR2, AGR3, CEACAM5, CD90, and SFRP4 in the digested sample using mass spectrometry.

19. The method of claim 18, further comprising:

measuring expression of CRISPS, CCL3, CEACAM6, IL24, MMP9, CXCL14, and POSTN in the digested sample using mass spectrometry.

20. A method of treating a subject with intermediate- or high-risk prostate cancer, comprising:

measuring expression of MMP9 in a sample obtained from a subject, wherein the expression is determined based on MMP9 protein concentration using at least one surrogate peptide of MMP9, wherein the surrogate peptide is at least one of FQTFEGDLK (SEQ ID NO: 41) or LGLGADVAQVTGALR (SEQ ID NO: 42);
measuring increased expression of MMP9 in the sample obtained from the subject as compared to a control representing expression of MMP9 expected in a sample from a subject who has low-risk prostate cancer; and
administering treatment for intermediate- or high-risk prostate cancer, thereby treating the subject.
Patent History
Publication number: 20180164329
Type: Application
Filed: Dec 12, 2017
Publication Date: Jun 14, 2018
Patent Grant number: 10725053
Applicants: ,
Inventors: Wei-Jun Qian (Richland, WA), Tujin Shi (Richland, WA), Alvin Y. Liu (Everett, WA)
Application Number: 15/839,635
Classifications
International Classification: G01N 33/68 (20060101); C12Q 1/37 (20060101); C12Q 1/533 (20060101);